1
|
Wei Y, Li R, Wang Y, Fu J, Liu J, Ma X. Nanomedicines Targeting Tumor Cells or Tumor-Associated Macrophages for Combinatorial Cancer Photodynamic Therapy and Immunotherapy: Strategies and Influencing Factors. Int J Nanomedicine 2024; 19:10129-10144. [PMID: 39381025 PMCID: PMC11460276 DOI: 10.2147/ijn.s466315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024] Open
Abstract
Immunotherapy is a promising cancer treatment because of its ability to sustainably enhance the natural immune response. However, the effects of multiple immunotherapies, including ICIs, are limited by resistance to these agents, immune-related adverse events, and a lack of reasonable therapeutic targets available at the right time and place. The tumor microenvironment (TME), which features tumor-associated macrophages (TAMs), plays a significant role in resistance owing to its hypoxic microenvironment and lack of blood vessels, resulting in cancer immune evasion. To enhance immunotherapy, photodynamic therapy (PDT) can increase innate and adaptive immune responses through immunogenic cell death (ICD) and improve the TME. Traditional photosensitizers (PSs) also include novel nanomedicines to precisely target tumor cells or TAMs. Here, we reviewed and summarized current strategies and possible influencing factors for nanomedicines for cancer photoimmunotherapy.
Collapse
Affiliation(s)
- Yuhao Wei
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Renwei Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Yusha Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Jiali Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu, People’s Republic of China
| | - Jifeng Liu
- Department of Otolaryngology Head and Neck Surgery/Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- State Key Laboratory of Intelligent Construction and Healthy Operation and Maintenance of Deep Underground Engineering, Sichuan University, Chengdu, People’s Republic of China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| |
Collapse
|
2
|
Hlapisi N, Songca SP, Ajibade PA. Capped Plasmonic Gold and Silver Nanoparticles with Porphyrins for Potential Use as Anticancer Agents-A Review. Pharmaceutics 2024; 16:1268. [PMID: 39458600 PMCID: PMC11510308 DOI: 10.3390/pharmaceutics16101268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/16/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Photothermal therapy (PTT) and photodynamic therapy (PDT) are potential cancer treatment methods that are minimally invasive with high specificity for malignant cells. Emerging research has concentrated on the application of metal nanoparticles encapsulated in porphyrin and their derivatives to improve the efficacy of these treatments. Gold and silver nanoparticles have distinct optical properties and biocompatibility, which makes them efficient materials for PDT and PTT. Conjugation of these nanoparticles with porphyrin derivatives increases their light absorption and singlet oxygen generation that create a synergistic effect that increases phototoxicity against cancer cells. Porphyrin encapsulation with gold or silver nanoparticles improves their solubility, stability, and targeted tumor delivery. This paper provides comprehensive review on the design, functionalization, and uses of plasmonic silver and gold nanoparticles in biomedicine and how they can be conjugated with porphyrins for synergistic therapeutic effects. Furthermore, it investigates this dual-modal therapy's potential advantages and disadvantages and offers perspectives for future prospects. The possibility of developing gold, silver, and porphyrin nanotechnology-enabled biomedicine for combination therapy is also examined.
Collapse
Affiliation(s)
| | | | - Peter A. Ajibade
- School of Chemistry and Physics, University of KwaZulu-Natal, Private Bag X01, Scottsville, Pietermaritzburg 3209, South Africa; (N.H.); (S.P.S.)
| |
Collapse
|
3
|
Hoshi R, Gorospe KA, Labouta HI, Azad T, Lee WL, Thu KL. Alternative Strategies for Delivering Immunotherapeutics Targeting the PD-1/PD-L1 Immune Checkpoint in Cancer. Pharmaceutics 2024; 16:1181. [PMID: 39339217 PMCID: PMC11434872 DOI: 10.3390/pharmaceutics16091181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
The programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) immune checkpoint constitutes an inhibitory pathway best known for its regulation of cluster of differentiation 8 (CD8)+ T cell-mediated immune responses. Engagement of PD-L1 with PD-1 expressed on CD8+ T cells activates downstream signaling pathways that culminate in T cell exhaustion and/or apoptosis. Physiologically, these immunosuppressive effects exist to prevent autoimmunity, but cancer cells exploit this pathway by overexpressing PD-L1 to facilitate immune escape. Intravenously (IV) administered immune checkpoint inhibitors (ICIs) that block the interaction between PD-1/PD-L1 have achieved great success in reversing T cell exhaustion and promoting tumor regression in various malignancies. However, these ICIs can cause immune-related adverse events (irAEs) due to off-tumor toxicities which limits their therapeutic potential. Therefore, considerable effort has been channeled into exploring alternative delivery strategies that enhance tumor-directed delivery of PD-1/PD-L1 ICIs and reduce irAEs. Here, we briefly describe PD-1/PD-L1-targeted cancer immunotherapy and associated irAEs. We then provide a detailed review of alternative delivery approaches, including locoregional (LDD)-, oncolytic virus (OV)-, nanoparticle (NP)-, and ultrasound and microbubble (USMB)-mediated delivery that are currently under investigation for enhancing tumor-specific delivery to minimize toxic off-tumor effects. We conclude with a commentary on key challenges associated with these delivery methods and potential strategies to mitigate them.
Collapse
Affiliation(s)
- Ryunosuke Hoshi
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| | - Kristyna A. Gorospe
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| | - Hagar I. Labouta
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
- Leslie Dan Faculty of Pharmacy, University of Toronto, St. George Campus, Toronto, ON M5S 3M2, Canada
- Biomedical Engineering, Faculty of Applied Science and Engineering, University of Toronto, St. George Campus, Toronto, ON M5S 3E2, Canada
| | - Taha Azad
- Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Health Campus, Sherbrooke, QC J1K 2R1, Canada;
- Research Center, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC J1J 3H5, Canada
| | - Warren L. Lee
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
- Biochemistry, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada
- Medicine and the Interdepartmental Division of Critical Care Medicine, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5B 1T8, Canada
| | - Kelsie L. Thu
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| |
Collapse
|
4
|
Li N, Cui S, Yang A, Xiao B, Cao Y, Yang X, Lin C. Sequence-dependent effects of hematoporphyrin derivatives (HPD) photodynamic therapy and cisplatin on lung adenocarcinoma cells. Photodiagnosis Photodyn Ther 2024; 47:104102. [PMID: 38679153 DOI: 10.1016/j.pdpdt.2024.104102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/07/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Hematoporphyrin derivatives (HPD)-Photodynamic therapy (PDT) in combination with cisplatin (DDP) is an effective anticancer strategy. However, whether the order of combination affects efficacy has not been studied. METHODS The human lung adenocarcinoma (LUAD) A549 cells were used as the study subjects. After A549 cells were treated with a single medication (PDT/DDP) or a sequential combination (PDT + DDP / DDP + PDT), the cell viability was assayed using the cell counting kit-8 method. Hoechst staining, Annexin-V/propidium iodide (PI) double staining, western blotting, and a real-time quantitative polymerase chain reaction (RT-qPCR) were performed to examine the mechanisms behind the combined effects. RESULTS A synergistic impact between HPD-PDT and DDP was found. The cell viability in the PDT+DDP group was significantly lower than in the DDP+PDT group. A significant apoptotic profile and a high apoptotic rate were seen in the PDT + DDP group. The western blot showed that the expression levels of Bcl2-associated x(Bax) and cleaved-poly ADP-ribose polymerase (PARP) increased, and those of B-cell lymphoma-2 (Bcl-2) and Caspase-9 decreased in the PDT + DDP group. At the same time, the RT-qPCR revealed the upregulation of Bax and PARP mRNA and the downregulation of Bcl-2 and Caspase-9 mRNA. CONCLUSION The order of the combination therapy (PDT + DDP / DDP + PDT) was important. The HPD-PDT followed by DDP significantly inhibited LUAD cell viability, which may be related to the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Nana Li
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Shichao Cui
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Aizhen Yang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Baohong Xiao
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yiwei Cao
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xiaohui Yang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Cunzhi Lin
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
5
|
Li F, Wang H, Ye T, Guo P, Lin X, Hu Y, Wei W, Wang S, Ma G. Recent Advances in Material Technology for Leukemia Treatments. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313955. [PMID: 38547845 DOI: 10.1002/adma.202313955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/11/2024] [Indexed: 04/13/2024]
Abstract
Leukemia is a widespread hematological malignancy characterized by an elevated white blood cell count in both the blood and the bone marrow. Despite notable advancements in leukemia intervention in the clinic, a large proportion of patients, especially acute leukemia patients, fail to achieve long-term remission or complete remission following treatment. Therefore, leukemia therapy necessitates optimization to meet the treatment requirements. In recent years, a multitude of materials have undergone rigorous study to serve as delivery vectors or direct intervention agents to bolster the effectiveness of leukemia therapy. These materials include liposomes, protein-based materials, polymeric materials, cell-derived materials, and inorganic materials. They possess unique characteristics and are applied in a broad array of therapeutic modalities, including chemotherapy, gene therapy, immunotherapy, radiotherapy, hematopoietic stem cell transplantation, and other evolving treatments. Here, an overview of these materials is presented, describing their physicochemical properties, their role in leukemia treatment, and the challenges they face in the context of clinical translation. This review inspires researchers to further develop various materials that can be used to augment the efficacy of multiple therapeutic modalities for novel applications in leukemia treatment.
Collapse
Affiliation(s)
- Feng Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huaiji Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tong Ye
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peilin Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoyun Lin
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yuxing Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
6
|
Zumaya ALV, Pavlíčková VS, Rimpelová S, Štějdířová M, Fulem M, Křížová I, Ulbrich P, Řezanka P, Hassouna F. PLGA-based nanocarriers for combined delivery of colchicine and purpurin 18 in cancer therapy: Multimodal approach employing cancer cell spheroids. Int J Pharm 2024; 657:124170. [PMID: 38679244 DOI: 10.1016/j.ijpharm.2024.124170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Improving the anticancer efficacy of chemotherapeutic drugs and photosensitizers requires innovative multifunctional nanoplatforms. This study introduces a chemo- and phototherapeutic drug delivery system (DDS) based on poly(lactide-co-glycolide) (PLGA) nanoparticles (NPs), both PEGylated and non-PEGylated, with a mean size of 200 ± 75 nm. Colchicine (Colch) and purpurin18 (P18) were co-encapsulated into these NPs, and their in vitro drug release profiles were investigated. The anticancer potential of these systems was evaluated across various cell lines (i.e., CaCo-2, PC-3, MCF-7, and MRC-5 cells), demonstrating enhanced NP uptake by cancer cells compared to free drugs. Co-administration of Colch and P18 in 2D and 3D cell line models exhibited a synergistic effect, harnessing both chemotherapeutic and photodynamic effects, leading to higher cancer cell elimination efficacy. This newly developed multifunctional DDS presents a promising platform for combined chemo- and photodynamic therapy in cancer treatment.
Collapse
Affiliation(s)
- Alma Lucia Villela Zumaya
- Faculty of Chemical Engineering, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Vladimíra Svobodová Pavlíčková
- Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Silvie Rimpelová
- Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic.
| | - Markéta Štějdířová
- Faculty of Chemical Engineering, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Michal Fulem
- Faculty of Chemical Engineering, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Ivana Křížová
- Faculty of Biotechnology, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Pavel Ulbrich
- Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Pavel Řezanka
- Faculty of Chemical Engineering, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Fatima Hassouna
- Faculty of Chemical Engineering, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic.
| |
Collapse
|
7
|
Jiang A, Wang T, Lu X, Tian Y, Jiang Z, Xu B, Zhang H, Fang W. Synthesis of Fe 3O 4core/MIL-100(Fe) shell nanocomposites for tumor chemo-ferroptosis combination therapy and MR imaging. Biomed Mater 2024; 19:025018. [PMID: 38215473 DOI: 10.1088/1748-605x/ad1dfe] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/12/2024] [Indexed: 01/14/2024]
Abstract
The application of both chemotherapy and ferrotherapy together has shown great potential in increasing the effectiveness of cancer treatment. To achieve such a combination, we herein have synthesized Fe3O4core/MIL-100(Fe) shell nanocomposites (FM) that can be used for tumor chemo-ferroptosis combination therapy. In these nanocomposites, the anticancer drug 10-hydroxycamptothecin (HCPT) and iron ions could be co-delivered into tumors. On one hand, the released HCPT molecules can enter the cell nucleus and bind with DNA, resulting in induction of tumor cell apoptosis. On the other hand, the iron ions could react with H2O2leading to the production of ROS through the Fenton reaction, thereby triggering tumor cell ferroptosis. Consequently, a superior antitumor effect was achieved through the combination of the apoptosis and ferroptosis. Additionally, the Fe3O4core endowed FM with high performance for magnetic resonance imaging, which further provided novel avenues for imaging guidance therapy. Therefore, we anticipate that application of these nanocomposites could have great potential in the field of tumor therapy.
Collapse
Affiliation(s)
- Antong Jiang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Teng Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, People's Republic of China
| | - Xiaoling Lu
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, People's Republic of China
| | - Yuxiang Tian
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, People's Republic of China
| | - Zihan Jiang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, People's Republic of China
| | - Bin Xu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, People's Republic of China
| | - Hanyuan Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Weijun Fang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, Anhui, People's Republic of China
| |
Collapse
|
8
|
Shaw PA, Klausen M, Bradley M. A dual action coumarin-camptothecin polymer for light responsive drug release and photodynamic therapy. Polym Chem 2024; 15:54-58. [PMID: 38174055 PMCID: PMC10758926 DOI: 10.1039/d3py01137b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024]
Abstract
A light-responsive polymer allowing the controlled release of camptothecin and the generation of reactive oxygen species (ROS) is reported. The polymer was prepared by controlled copolymerisation of water-soluble N,N-dimethyl acrylamide with a bromocoumarin methacrylate monomer. The lipophilic chemotherapy agent camptothecin was caged onto the coumarin unit via a photo-cleavable carbonate ester enabling light-triggered cargo release. The polymer showed good biocompatibility in the dark, and high cancer cell killing activity mediated both by the photo-release of camptothecin and ROS generation.
Collapse
Affiliation(s)
- Paige A Shaw
- EaStCHEM School of Chemistry, University of Edinburgh David Brewster Road EH9 3FJ Edinburgh UK
| | - Maxime Klausen
- EaStCHEM School of Chemistry, University of Edinburgh David Brewster Road EH9 3FJ Edinburgh UK
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London London SW7 2AZ UK
| | - Mark Bradley
- EaStCHEM School of Chemistry, University of Edinburgh David Brewster Road EH9 3FJ Edinburgh UK
- Precision Healthcare University Research Institute, Queen Mary University of London Empire House London E1 1HH UK
| |
Collapse
|
9
|
Shi E, Shan T, Wang H, Mao L, Liang Y, Cao M, Wu Q, Li C, Wang Y, Wang Y. A Bacterial Nanomedicine Combines Photodynamic-Immunotherapy and Chemotherapy for Enhanced Treatment of Oral Squamous Cell Carcinoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2304014. [PMID: 37653616 DOI: 10.1002/smll.202304014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/08/2023] [Indexed: 09/02/2023]
Abstract
Bacterial therapy is an emerging hotspot in tumor immunotherapy, which can initiate antitumor immune activation through multiple mechanisms. Porphyromonas gingivalis (Pg), a pathogenic bacterium inhabiting the oral cavity, contains a great deal of pathogen associated molecular patterns that can activate various innate immune cells to promote antitumor immunity. Owing to the presence of protoporphyrin IX (PpIX), Pg is also an excellent photosensitizer for photodynamic therapy (PDT) via the in situ generation of reactive oxygen species. This study reports a bacterial nanomedicine (nmPg) fabricated from Pg through lysozyme degradation, ammonium chloride lysis, and nanoextrusion, which has potent PDT and immune activation performances for oral squamous cell carcinoma (OSCC) treatment. To further promote the tumoricidal efficacy, a commonly used chemotherapeutic drug doxorubicin (DOX) is efficiently encapsulated into nmPg through a simple incubation method. nmPg/DOX thus prepared exhibits significant synergistic effects on inhibiting the growth and metastasis of OSCC both in vitro and in vivo via photodynamic-immunotherapy and chemotherapy. In summary, this work develops a promising bacterial nanomedicine for enhanced treatment of OSCC.
Collapse
Affiliation(s)
- Enyu Shi
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Tianhe Shan
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| | - Hanping Wang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Lujia Mao
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Yanjie Liang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Mingxin Cao
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Qiqi Wu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Changyi Li
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Yue Wang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
| | - Yinsong Wang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
10
|
Bhattacharya D, Mukhopadhyay M, Shivam K, Tripathy S, Patra R, Pramanik A. Recent developments in photodynamic therapy and its application against multidrug resistant cancers. Biomed Mater 2023; 18:062005. [PMID: 37827172 DOI: 10.1088/1748-605x/ad02d4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/12/2023] [Indexed: 10/14/2023]
Abstract
Recently, photodynamic therapy (PDT) has received a lot of attention for its potential use in cancer treatment. It enables the therapy of a multifocal disease with the least amount of tissue damage. The most widely used prodrug is 5-aminolevulinic acid, which undergoes heme pathway conversion to protoporphyrin IX, which acts as a photosensitizer (PS). Additionally, hematoporphyrin, bacteriochlorin, and phthalocyanine are also studied for their therapeutic potential in cancer. Unfortunately, not every patient who receives PDT experiences a full recovery. Resistance to different anticancer treatments is commonly observed. A few of the resistance mechanisms by which cancer cells escape therapeutics are genetic factors, drug-drug interactions, impaired DNA repair pathways, mutations related to inhibition of apoptosis, epigenetic pathways, etc. Recently, much research has been conducted to develop a new generation of PS based on nanomaterials that could be used to overcome cancer cells' multidrug resistance (MDR). Various metal-based, polymeric, lipidic nanoparticles (NPs), dendrimers, etc, have been utilized in the PDT application against cancer. This article discusses the detailed mechanism by which cancer cells evolve towards MDR as well as recent advances in PDT-based NPs for use against multidrug-resistant cancers.
Collapse
Affiliation(s)
- Debalina Bhattacharya
- Department of Microbiology, Maulana Azad College, Kolkata, West Bengal 700013, India
| | - Mainak Mukhopadhyay
- Department of Biotechnology, JIS University, Kolkata, West Bengal 700109, India
| | - Kumar Shivam
- Amity Institute of Click Chemistry Research & Studies, Amity University, Noida 201301, India
| | - Satyajit Tripathy
- Department of Pharmacology, University of Free State, Bloemfontein, Free State, 9301, South Africa
- Amity Institute of Allied Health Science, Amity University, Noida 201301, India
| | - Ranjan Patra
- Amity Institute of Click Chemistry Research & Studies, Amity University, Noida 201301, India
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Arindam Pramanik
- School of Medicine, University of Leeds, Leeds, LS9 7TF, United Kingdom
- Amity Institute of Biotechnology, Amity University, Noida 201301, India
| |
Collapse
|
11
|
Laneri F, Seggio M, Parisi C, Béni S, Fraix A, Malanga M, Sortino S. Mixed β-γ-Cyclodextrin Branched Polymer with Multiple Photo-Chemotherapeutic Cargos. ACS APPLIED POLYMER MATERIALS 2023; 5:7918-7926. [PMID: 37854303 PMCID: PMC10580695 DOI: 10.1021/acsapm.3c01157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/18/2023] [Indexed: 10/20/2023]
Abstract
The achievement of biocompatible platforms for multimodal therapies is one of the major challenges in the burgeoning field of nanomedicine. Here, we report on a mixed β- and γ-cyclodextrin-based branched polymeric material (βγCD-NOPD) covalently integrating a nitric oxide (NO) photodonor (NOPD) within its macromolecular scaffold, and its supramolecular ensemble with a singlet oxygen (1O2) photosensitizer (PS) Zn(II) phthalocyanine (ZnPc) and the chemodrug Lenvatinib (LVB). This polymer is highly water-soluble and generates NO under visible blue light stimuli with an efficiency of more than 1 order of magnitude higher than that of the single NOPD. The PS, which in an aqueous solution is aggregated and non-photoresponsive, can be entangled in the polymeric network as a photoresponsive monomeric species. In addition, the poorly water-soluble LVB can be co-encapsulated within the polymeric host, which increases the drug solubility by more than 30-fold compared to the free drug and more than 2-fold compared with a similar branched polymer containing only βCD units. The supramolecular nanoensemble, ca. 15 nm in diameter, retains well the photochemical properties of both the NOPD and PS, which can operate in parallel under light stimuli of different energies. Irradiation with blue and red light results in the photogeneration of NO and 1O2 associated with red fluorescence emission, without inducing any photodegradation of LVB. This result is not trivial and is due to the absence of significant, mutual interactions between the NOPD, the PS and LVB both in the ground and excited states, despite these components are confined in the same host. The proposed polymeric nanoplatform may represent a potential trimodal nanomedicine for biomedical research studies, since it combines the double photodynamic action of NO and 1O2, two species that do not suffer multidrug resistance, with the therapeutic activity of a conventional chemodrug.
Collapse
Affiliation(s)
- Francesca Laneri
- PhotoChemLab,
Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| | - Mimimorena Seggio
- PhotoChemLab,
Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| | - Cristina Parisi
- PhotoChemLab,
Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| | - Szabolcs Béni
- Department
of Pharmacognosy, Semmelweis University, I-1085 Budapest, Hungary
| | - Aurore Fraix
- PhotoChemLab,
Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| | - Milo Malanga
- CycloLab,
Cyclodextrin R&D Ltd., I-1097 Budapest, Hungary
| | - Salvatore Sortino
- PhotoChemLab,
Department of Drug and Health Sciences, University of Catania, I-95125 Catania, Italy
| |
Collapse
|
12
|
Fan D, Cao Y, Cao M, Wang Y, Cao Y, Gong T. Nanomedicine in cancer therapy. Signal Transduct Target Ther 2023; 8:293. [PMID: 37544972 PMCID: PMC10404590 DOI: 10.1038/s41392-023-01536-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 08/08/2023] Open
Abstract
Cancer remains a highly lethal disease in the world. Currently, either conventional cancer therapies or modern immunotherapies are non-tumor-targeted therapeutic approaches that cannot accurately distinguish malignant cells from healthy ones, giving rise to multiple undesired side effects. Recent advances in nanotechnology, accompanied by our growing understanding of cancer biology and nano-bio interactions, have led to the development of a series of nanocarriers, which aim to improve the therapeutic efficacy while reducing off-target toxicity of the encapsulated anticancer agents through tumor tissue-, cell-, or organelle-specific targeting. However, the vast majority of nanocarriers do not possess hierarchical targeting capability, and their therapeutic indices are often compromised by either poor tumor accumulation, inefficient cellular internalization, or inaccurate subcellular localization. This Review outlines current and prospective strategies in the design of tumor tissue-, cell-, and organelle-targeted cancer nanomedicines, and highlights the latest progress in hierarchical targeting technologies that can dynamically integrate these three different stages of static tumor targeting to maximize therapeutic outcomes. Finally, we briefly discuss the current challenges and future opportunities for the clinical translation of cancer nanomedicines.
Collapse
Affiliation(s)
- Dahua Fan
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, China.
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| | - Yongkai Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Meiqun Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Yajun Wang
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, China
| | | | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
13
|
Zhou H, Liao Y, Han X, Chen DS, Hong X, Zhou K, Jiang X, Xiao Y, Shi J. ROS-Responsive Nanoparticle Delivery of mRNA and Photosensitizer for Combinatorial Cancer Therapy. NANO LETTERS 2023; 23:3661-3668. [PMID: 37093620 DOI: 10.1021/acs.nanolett.2c03784] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Messenger RNA (mRNA) therapy has shown tremendous potential for different diseases including cancer. While mRNA has been extensively used in cancer vaccine development as antigen or in cancer immunotherapy as immunomodulatory agent, the combination of mRNA therapy with photodynamic therapy has not been explored in cancer treatment. Herein, we report a reactive oxygen species (ROS)-responsive polymeric nanoparticle (NP) platform for first-in-field codelivery of mRNA and photosensitizer for effective cancer treatment. We developed ROS-responsive oligomer-based polymeric NPs and applied them to test a combination of p53 mRNA and indocyanine green (ICG). The ROS-triggered disassembly of the NPs could promote mRNA translation efficiency, whereby p53 expression induced apoptosis of lung tumor cells. Meanwhile, the released ICG could lead to generation of ROS under 808 nm laser irradiation to induce photodynamic therapy. The NP codelivery of p53 mRNA and ICG demonstrated an effective and safe anti-tumor effect in a lung cancer model.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Yuqin Liao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Xiangfei Han
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Dean Shuailin Chen
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Xuechuan Hong
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Shenzhen Institute of Wuhan University, Shenzhen 518057, China
| | - Kun Zhou
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115 United States
| | - Xingya Jiang
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yuling Xiao
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
14
|
Mušković M, Pokrajac R, Malatesti N. Combination of Two Photosensitisers in Anticancer, Antimicrobial and Upconversion Photodynamic Therapy. Pharmaceuticals (Basel) 2023; 16:613. [PMID: 37111370 PMCID: PMC10143496 DOI: 10.3390/ph16040613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/12/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
Photodynamic therapy (PDT) is a special form of phototherapy in which oxygen is needed, in addition to light and a drug called a photosensitiser (PS), to create cytotoxic species that can destroy cancer cells and various pathogens. PDT is often used in combination with other antitumor and antimicrobial therapies to sensitise cells to other agents, minimise the risk of resistance and improve overall outcomes. Furthermore, the aim of combining two photosensitising agents in PDT is to overcome the shortcomings of the monotherapeutic approach and the limitations of individual agents, as well as to achieve synergistic or additive effects, which allows the administration of PSs in lower concentrations, consequently reducing dark toxicity and preventing skin photosensitivity. The most common strategies in anticancer PDT use two PSs to combine the targeting of different organelles and cell-death mechanisms and, in addition to cancer cells, simultaneously target tumour vasculature and induce immune responses. The use of PDT with upconversion nanoparticles is a promising approach to the treatment of deep tissues and the goal of using two PSs is to improve drug loading and singlet oxygen production. In antimicrobial PDT, two PSs are often combined to generate various reactive oxygen species through both Type I and Type II processes.
Collapse
Affiliation(s)
| | | | - Nela Malatesti
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia; (M.M.); (R.P.)
| |
Collapse
|
15
|
Bhattacharya T, Shin GH, Kim JT. Carbon Dots: Opportunities and Challenges in Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15031019. [PMID: 36986879 PMCID: PMC10059251 DOI: 10.3390/pharmaceutics15031019] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Recently, carbon dots (CDs) have been actively studied and reported for their various properties. In particular, the specific characteristics of carbon dots have been considered as a possible technique for cancer diagnosis and therapy. This is also a cutting-edge technology that offers fresh ideas for treating various disorders. Though carbon dots are still in their infancy and have not yet shown their value to society, their discovery has already resulted in some noteworthy advancements. The application of CDs indicates conversion in natural imaging. Photography using CDs has demonstrated extraordinary appropriateness in bio-imaging, the discovery of novel drugs, the delivery of targeted genes, bio-sensing, photodynamic therapy, and diagnosis. This review seeks to provide a comprehensive understanding of CDs, including their benefits, characteristics, applications, and mode of action. In this overview, many CD design strategies will be highlighted. In addition, we will discuss numerous studies on cytotoxic testing to demonstrate the safety of CDs. The current study will address the production method, mechanism, ongoing research, and application of CDs in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Tanima Bhattacharya
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Gye Hwa Shin
- Department of Food and Nutrition, Kunsan National University, Gunsan 54150, Republic of Korea
- Correspondence: (G.H.S.); (J.T.K.)
| | - Jun Tae Kim
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
- BioNanocomposite Research Center, Kyung Hee University, Seoul 02447, Republic of Korea
- Correspondence: (G.H.S.); (J.T.K.)
| |
Collapse
|
16
|
Zhang H, Wang S, Sun M, Cui Y, Xing J, Teng L, Xi Z, Yang Z. Exosomes as smart drug delivery vehicles for cancer immunotherapy. Front Immunol 2023; 13:1093607. [PMID: 36733388 PMCID: PMC9888251 DOI: 10.3389/fimmu.2022.1093607] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Exosomes (Exos) as drug delivery vehicles have been widely used for cancer immunotherapy owing to their good biocompatibility, low toxicity, and low immunogenicity. Some Exos-based cancer immunotherapy strategies such as tuning of immunosuppressive tumor microenvironment, immune checkpoint blockades, and cancer vaccines have also been investigated in recent years, which all showed excellent therapeutic effects for malignant tumor. Furthermore, some Exos-based drug delivery systems (DDSs) for cancer immunotherapy have also undergone clinic trails, indicating that Exos are a promising drug delivery carrier. In this review, in order to promote the development of Exos-based DDSs in cancer immunotherapy, the biogenesis and composition of Exos, and Exos as drug delivery vehicles for cancer immunotherapy are summarized. Meanwhile, their clinical translation and challenges are also discussed. We hope this review will provide a good guidance for Exos as drug delivery vehicles for cancer immunotherapy.
Collapse
Affiliation(s)
- Huan Zhang
- School of Life Sciences, Jilin University, Changchun, China
| | - Simiao Wang
- School of Life Sciences, Jilin University, Changchun, China
| | - Man Sun
- School of Life Sciences, Jilin University, Changchun, China
| | - Yaxin Cui
- School of Life Sciences, Jilin University, Changchun, China
| | - Jianming Xing
- School of Life Sciences, Jilin University, Changchun, China
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun, China
| | - Zhifang Xi
- School of Horticulture and Food, Guangdong Eco-Engineering Polytechnic, Guangzhou, China,*Correspondence: Zhifang Xi, ; Zhaogang Yang,
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun, China,*Correspondence: Zhifang Xi, ; Zhaogang Yang,
| |
Collapse
|
17
|
Development of a Polymersome-Based Nanomedicine for Chemotherapeutic and Sonodynamic Combination Therapy. Int J Mol Sci 2023; 24:ijms24021194. [PMID: 36674707 PMCID: PMC9864053 DOI: 10.3390/ijms24021194] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
In anticancer therapy, combination therapy has been suggested as an alternative to the insufficient therapeutic efficacy of single therapy. Among combination therapies, combination chemo- and photodynamic therapy are actively investigated. However, photodynamic therapy shows a limitation in the penetration depth of the laser. Therefore, sonodynamic therapy (SDT), using ultrasound instead of a laser as a trigger, is an upcoming strategy for deep tumors. Additionally, free drugs are easily degraded by enzymes, have difficulty in reaching the target site, and show side effects after systemic administration; therefore, the development of drug delivery systems is desirable for sufficient drug efficacy for combination therapy. However, nanocarriers, such as microbubbles, and albumin nanoparticles, are unstable in the body and show low drug-loading efficiency. Here, we propose polylactide (PLA)-poly (ethylene glycol) (PEG) polymersomes (PLs) with a high drug loading rate of doxorubicin (DOX) and verteporfin (VP) for effective combination therapy in both in vitro and in vivo experiments. The cellular uptake efficiency and cytotoxicity test results of VP-DOX-PLs were higher than that of single therapy. Moreover, in vivo biodistribution showed the accumulation of the VP-DOX-PLs in tumor regions. Therefore, VP-DOX-PLs showed more effective anticancer efficacy than either single therapy in vivo. These results suggest that the combination therapy of SDT and chemotherapy could show novel anticancer effects using VP-DOX-PLs.
Collapse
|
18
|
A pH-Responsive Drug Delivery System Based on Conjugated Polymer for Effective Synergistic Chemo-/Photodynamic Therapy. Molecules 2023; 28:molecules28010399. [PMID: 36615594 PMCID: PMC9823741 DOI: 10.3390/molecules28010399] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 01/05/2023] Open
Abstract
Stimuli-responsive drug release and photodynamic therapy (PDT) have aroused extensive attention for their enormous potential in antitumor treatment. pH-responsive drug delivery systems (PFE-DOX-1 and PFE-DOX-2) based on water-soluble conjugated polymers were constructed in this work for high-performance synergistic chemo-/PDT therapy, in which the anticancer drug doxorubicin (DOX) is covalently attached to the side chains of the conjugated polymers via acid-labile imine and acylhydrazone bonds. Concurrently, the intense fluorescence of poly(fluorene-co-ethynylene) (PFE) is effectively quenched due to the energy/electron transfer (ET) between the PFE-conjugated backbone and DOX. Effective pH-responsive drug release from PFE-DOX-2 is achieved by the cleavage of acylhydrazone linkages in the acidic tumor intracellular microenvironment. Additionally, the drug release process can be monitored by the recovered fluorescence of conjugated polymers. Furthermore, the conjugated polymers can produce reactive oxygen species (ROS) under light irradiation after drug release in an acidic environment, which prevents possible phototoxicity to normal tissues. It is noted that PFE-DOX-2 demonstrates remarkable antitumor cell performance, which is attributed to its efficient cell uptake and powerful synergistic chemo-/PDT therapeutic effectiveness. This report thus provides a promising strategy for in vivo anticancer treatment with the construction of a stimuli-responsive multifunctional drug delivery system.
Collapse
|
19
|
Prodrug and Glucose Oxidase Coloaded Photodynamic Hydrogels for Combinational Therapy of Melanoma. ACS Biomater Sci Eng 2022; 8:4886-4895. [PMID: 36278808 DOI: 10.1021/acsbiomaterials.2c00992] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
With the advantages of high safety and selectivity, photodynamic therapy (PDT) has been widely used for cancer treatments, while the anticancer efficacy is often limited because of its relying on oxygen concentrations. Therefore, sole PDT fails to achieve the desired therapeutic effect for hypoxic tumors. To address this issue, we herein report the construction of prodrug and glucose oxidase (GOx) coloaded alginate (ALG) hydrogels for PDT-combined chemotherapy of melanoma. The hydrogels are in situ formed in tumor sites after injection of ALG solution containing semiconducting polymer nanoparticles, hypoxia-responsive prodrug tirapazamine (TPZ), and GOx, which is based on chelation of ALG by endogenous Ca2+. Due to the presence of semiconducting polymer nanoparticles acting as photosensitizers, the hydrogels mediate PDT to produce singlet oxygen (1O2) for directly killing tumor cells, in which oxygen is consumed to create a more hypoxic tumor microenvironment. Moreover, the loaded GOx within hydrogels can deplete oxygen to further aggravate tumor hypoxia. As such, TPZ is effectively activated by hypoxia to cause cancer cell death via chemotherapy. Thus, the hydrogels with laser irradiation achieve a combinational action of PDT with chemotherapy to almost completely eradicate tumors, leading to a much higher therapeutic efficacy relative to sole PDT. This study will provide a promising injectable hydrogel platform for effective treatments of cancer.
Collapse
|
20
|
Chapla R, Huynh KT, Schutt CE. Microbubble–Nanoparticle Complexes for Ultrasound-Enhanced Cargo Delivery. Pharmaceutics 2022; 14:pharmaceutics14112396. [PMID: 36365214 PMCID: PMC9698658 DOI: 10.3390/pharmaceutics14112396] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 11/09/2022] Open
Abstract
Targeted delivery of therapeutics to specific tissues is critically important for reducing systemic toxicity and optimizing therapeutic efficacy, especially in the case of cytotoxic drugs. Many strategies currently exist for targeting systemically administered drugs, and ultrasound-controlled targeting is a rapidly advancing strategy for externally-stimulated drug delivery. In this non-invasive method, ultrasound waves penetrate through tissue and stimulate gas-filled microbubbles, resulting in bubble rupture and biophysical effects that power delivery of attached cargo to surrounding cells. Drug delivery capabilities from ultrasound-sensitive microbubbles are greatly expanded when nanocarrier particles are attached to the bubble surface, and cargo loading is determined by the physicochemical properties of the nanoparticles. This review serves to highlight and discuss current microbubble–nanoparticle complex component materials and designs for ultrasound-mediated drug delivery. Nanocarriers that have been complexed with microbubbles for drug delivery include lipid-based, polymeric, lipid–polymer hybrid, protein, and inorganic nanoparticles. Several schemes exist for linking nanoparticles to microbubbles for efficient nanoparticle delivery, including biotin–avidin bridging, electrostatic bonding, and covalent linkages. When compared to unstimulated delivery, ultrasound-mediated cargo delivery enables enhanced cell uptake and accumulation of cargo in target organs and can result in improved therapeutic outcomes. These ultrasound-responsive delivery complexes can also be designed to facilitate other methods of targeting, including bioactive targeting ligands and responsivity to light or magnetic fields, and multi-level targeting can enhance therapeutic efficacy. Microbubble–nanoparticle complexes present a versatile platform for controlled drug delivery via ultrasound, allowing for enhanced tissue penetration and minimally invasive therapy. Future perspectives for application of this platform are also discussed in this review.
Collapse
Affiliation(s)
- Rachel Chapla
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
| | - Katherine T. Huynh
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
| | - Carolyn E. Schutt
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
- Correspondence:
| |
Collapse
|
21
|
Wang Y, Zhang Y, Zhang X, Zhang Z, She J, Wu D, Gao W. High Drug-Loading Nanomedicines for Tumor Chemo-Photo Combination Therapy: Advances and Perspectives. Pharmaceutics 2022; 14:pharmaceutics14081735. [PMID: 36015361 PMCID: PMC9415722 DOI: 10.3390/pharmaceutics14081735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 11/28/2022] Open
Abstract
The combination of phototherapy and chemotherapy (chemo−photo combination therapy) is an excellent attempt for tumor treatment. The key requirement of this technology is the high drug-loading nanomedicines, which can load either chemotherapy drugs or phototherapy agents at the same nanomedicines and simultaneously deliver them to tumors, and play a multimode therapeutic role for tumor treatment. These nanomedicines have high drug-loading efficiency (>30%) and good tumor combination therapeutic effect with important clinical application potential. Although there are many reports of high drug-loading nanomedicines for tumor therapy at present, systematic analyses on those nanomedicines remain lacking and a comprehensive review is urgently needed. In this review, we systematically analyze the current status of developed high drug-loading nanomedicines for tumor chemo−photo combination therapy and summarize their types, methods, drug-loading properties, in vitro and in vivo applications. The shortcomings of the existing high drug-loading nanomedicines for tumor chemo−photo combination therapy and the possible prospective development direction are also discussed. We hope to attract more attention for researchers in different academic fields, provide new insights into the research of tumor therapy and drug delivery system and develop these nanomedicines as the useful tool for tumor chemo−photo combination therapy in the future.
Collapse
Affiliation(s)
- Ya Wang
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an 710061, China
| | - Yujie Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an 710061, China
| | - Xiaojiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an 710061, China
| | - Zhe Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an 710061, China
| | - Junjun She
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an 710061, China
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, 277 West Yanta Road, Xi’an 710061, China
- Correspondence: (J.S.); (D.W.); (W.G.)
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
- Correspondence: (J.S.); (D.W.); (W.G.)
| | - Wei Gao
- Department of Anesthesiology & Center for Brain Science & Center for Translational Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Correspondence: (J.S.); (D.W.); (W.G.)
| |
Collapse
|
22
|
Mills H, Acquah R, Tang N, Cheung L, Klenk S, Glassen R, Pirson M, Albert A, Hoang DT, Van TN. A Critical Scrutiny on Liposomal Nanoparticles Drug Carriers as Modelled by Topotecan Encapsulation and Release in Treating Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:7702512. [PMID: 35983007 PMCID: PMC9381203 DOI: 10.1155/2022/7702512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/10/2022] [Accepted: 07/27/2022] [Indexed: 11/18/2022]
Abstract
The medical field is looking for drugs and/or ways of delivering drugs without harming patients. A number of severe drug side effects are reported, such as acute kidney injury (AKI), hepatotoxicity, skin rash, and so on. Nanomedicine has come to the rescue. Liposomal nanoparticles have shown great potential in loading drugs, and delivering drugs to specific targeted sites, hence achieving a needed bioavailability and steady state concentration, which is achieved by a controlled drug release ability by the nanoparticles. The liposomal nanoparticles can be conjugated to cancer receptor tags that give the anticancer-loaded nanoparticles specificity to deliver anticancer agents only at cancerous sites, hence circumventing destruction of normal cells. Also, the particles are biocompatible. The drugs are shielded by attack from the liver and other cytochrome P450 enzymes before reaching the desired sites. The challenge, however, is that the drug release is slow by these nanoparticles on their own. Scientists then came up with several ways to enhance drug release. Magnetic fields, UV light, infrared light, and so on are amongst the enhancers used by scientists to potentiate drug release from nanoparticles. In this paper, synthesis of liposomal nanoparticle formulations (liposomal-quantum dots (L-QDs), liposomal-quantum dots loaded with topotecan (L-QD-TPT)) and their analysis (cytotoxicity, drug internalization, loading efficiency, drug release rate, and the uptake of the drug and nanoparticles by the HeLa cells) are discussed.
Collapse
Affiliation(s)
- Hilla Mills
- Department of Medical Science, University for Development, Accra, Ghana
| | - Ronald Acquah
- Department of Medical Science, University for Development, Accra, Ghana
| | - Nova Tang
- RD Lab, The Hospital Institute for Hebal Research, Toluca, MEX 50200, Mexico
| | - Luke Cheung
- RD Lab, The Hospital Institute for Hebal Research, Toluca, MEX 50200, Mexico
| | - Susanne Klenk
- Research Institution of Clinical Biomedicine, Hospital University Medical Centre, Ulm 89000, Germany
| | - Ronald Glassen
- Research Institution of Clinical Biomedicine, Hospital University Medical Centre, Ulm 89000, Germany
| | - Magali Pirson
- Industrial Research Group, International College of Science and Technology, Route de Lennik 800, CP 590, Brussels 1070, Belgium
| | - Alain Albert
- Industrial Research Group, International College of Science and Technology, Route de Lennik 800, CP 590, Brussels 1070, Belgium
| | | | | |
Collapse
|
23
|
Sargazi S, ER S, Sacide Gelen S, Rahdar A, Bilal M, Arshad R, Ajalli N, Farhan Ali Khan M, Pandey S. Application of titanium dioxide nanoparticles in photothermal and photodynamic therapy of cancer: An updated and comprehensive review. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Choi J, Sun IC, Sook Hwang H, Yeol Yoon H, Kim K. Light-triggered photodynamic nanomedicines for overcoming localized therapeutic efficacy in cancer treatment. Adv Drug Deliv Rev 2022; 186:114344. [PMID: 35580813 DOI: 10.1016/j.addr.2022.114344] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/27/2022] [Accepted: 05/09/2022] [Indexed: 12/14/2022]
Abstract
Photodynamic nanomedicines have significantly enhanced the therapeutic efficacy of photosensitizers (PSs) by overcoming critical limitations of PSs such as poor water solubility and low tumor accumulation. Furthermore, functional photodynamic nanomedicines have enabled overcoming oxygen depletion during photodynamic therapy (PDT) and tissue light penetration limitation by supplying oxygen or upconverting light in targeted tumor tissues, resulting in providing the potential to overcome biological therapeutic barriers of PDT. Nevertheless, their localized therapeutic effects still remain a huddle for the effective treatment of metastatic- or recurrent tumors. Recently, newly designed photodynamic nanomedicines and their combination chemo- or immune checkpoint inhibitor therapy enable the systemic treatment of various metastatic tumors by eliciting antitumor immune responses via immunogenic cell death (ICD). This review introduces recent advances in photodynamic nanomedicines and their applications, focusing on overcoming current limitations. Finally, the challenges and future perspectives of the clinical translation of photodynamic nanomedicines in cancer PDT are discussed.
Collapse
Affiliation(s)
- Jiwoong Choi
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea; Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - In-Cheol Sun
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hee Sook Hwang
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Republic of Korea
| | - Hong Yeol Yoon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea.
| | - Kwangmeyung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea; Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea.
| |
Collapse
|
25
|
Su R, Zhang X, Peng Q, Wang W. Self-assembling porphyrin conjugate-carboplatin(IV) prodrug nanoparticles for enhancing high efficacy nasopharyngeal cancer and low systemic toxicity. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:1828-1844. [PMID: 35686461 DOI: 10.1080/09205063.2022.2087275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Nanomedicine has developed as a potential technique for successful cancer therapy. A simple supramolecular self-assembly process is a helpful strategy for generating carrier-free nanodrugs. Mixing photodynamic treatment with chemotherapy has been sought to obtain a high therapeutic impact. In this study, we effectively construct a nanocarrier (CD-Por-PEG: Ada-CPT-Pt(IV)) combined with Carboplatin prodrug (Ada-CPT-Pt(IV)) and photosensitizer porphyrin (CD-Por-PEG) by host-guest interactions to accomplish stimuli-response combination treatment. Supported by greater spatial control of the binding ratio among host-guest molecules, Carboplatin and porphyrin were independently altered with β-cyclodextrin and adamantane to produce the amphiphilic host-guest combination for sequential self-assembly into therapeutic nanoparticles. The colloidal stability of the produced CD-Por-PEG: Ada-CPT-Pt(IV)-NPs was excellent, with an average hydrodynamic diameter of ∼170 nm. The microscopy images showed that CD-Por-PEG: Ada-CPT-Pt(IV) could aggregate cells and generate ROS after light irradiation (630 nm). Monotherapy had a cytotoxicity three times greater than the CD-Por-PEG: Ada-CPT-Pt(IV) nanoparticles. Studies in mice carrying SUNE1 nasopharyngeal tumours showed that nanoparticles effectively suppressed tumour development without causing systemic damage in this examination. The current self-assembly nanosystem makes precise control over the photosensitizer and drug loading possible ratio. It reduces the systemic adverse toxicity issues of drugs carrier, making this system ideal for nasopharyngeal cancer treatment.
Collapse
Affiliation(s)
- Renjie Su
- Department of Otorhinolaryngology, The First People's Hospital of Wenling, Wenling, China
| | - Xiangwen Zhang
- Department of Otorhinolaryngology, The First People's Hospital of Wenling, Wenling, China
| | - Qianhua Peng
- Department of Otorhinolaryngology, The First People's Hospital of Wenling, Wenling, China
| | - Wenbin Wang
- Department of Otorhinolaryngology, The First People's Hospital of Wenling, Wenling, China
| |
Collapse
|
26
|
Li CH, Chang YC, Hsiao M, Chan MH. Ultrasound and Nanomedicine for Cancer-Targeted Drug Delivery: Screening, Cellular Mechanisms and Therapeutic Opportunities. Pharmaceutics 2022; 14:1282. [PMID: 35745854 PMCID: PMC9229768 DOI: 10.3390/pharmaceutics14061282] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 12/02/2022] Open
Abstract
Cancer is a disease characterized by abnormal cell growth. According to a report published by the World Health Organization (WHO), cancer is the second leading cause of death globally, responsible for an estimated 9.6 million deaths in 2018. It should be noted that ultrasound is already widely used as a diagnostic procedure for detecting tumorigenesis. In addition, ultrasound energy can also be utilized effectively for treating cancer. By filling the interior of lipospheres with gas molecules, these particles can serve both as contrast agents for ultrasonic imaging and as delivery systems for drugs such as microbubbles and nanobubbles. Therefore, this review aims to describe the nanoparticle-assisted drug delivery system and how it can enhance image analysis and biomedicine. The formation characteristics of nanoparticles indicate that they will accumulate at the tumor site upon ultrasonic imaging, in accordance with their modification characteristics. As a result of changing the accumulation of materials, it is possible to examine the results by comparing images of other tumor cell lines. It is also possible to investigate ultrasound images for evidence of cellular effects. In combination with a precision ultrasound imaging system, drug-carrying lipospheres can precisely track tumor tissue and deliver drugs to tumor cells to enhance the ability of this nanocomposite to treat cancer.
Collapse
Affiliation(s)
- Chien-Hsiu Li
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
| | - Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan;
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Hsien Chan
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
| |
Collapse
|
27
|
Chen N, He Y, Zang M, Zhang Y, Lu H, Zhao Q, Wang S, Gao Y. Approaches and materials for endocytosis-independent intracellular delivery of proteins. Biomaterials 2022; 286:121567. [DOI: 10.1016/j.biomaterials.2022.121567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022]
|
28
|
Cacaccio JC, Durrani FA, Missert JR, Pandey RK. Photodynamic Therapy in Combination with Doxorubicin Is Superior to Monotherapy for the Treatment of Lung Cancer. Biomedicines 2022; 10:857. [PMID: 35453607 PMCID: PMC9024488 DOI: 10.3390/biomedicines10040857] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/29/2022] [Accepted: 04/01/2022] [Indexed: 02/01/2023] Open
Abstract
We have previously shown that a radioactive (123I)-analog of methyl 3-(1'-(iodobexyloxy) ethyl-3-devinylpyropheophorbide-a (PET-ONCO), derived from chlorophyll-a can be used for positron emission tomography (PET) imaging of a variety of tumors, including those where 18F-FDG shows limitations. In this study, the photodynamic therapy (PDT) efficacy of the corresponding non-radioactive photosensitizer (PS) was investigated in a variety of tumor types (NSCLC, SCC, adenocarcinoma) derived from lung cancer patients in mice tumor models. The in vitro and in vivo efficacy was also investigated in combination with doxorubicin, and a significantly enhanced long-term tumor response was observed. The toxicity and toxicokinetic profile of the iodinated PS was also evaluated in male and female Sprague-Dawley rats and Beagle dog at variable doses (single intravenous injections) to assess reversibility or latency of any effects over a 28-day dose free period. The no-observed-adverse-effect (NOAEL) of the PS was considered to be 6.5 mg/kg for male and female rats, and for dogs, 3.45 mg/kg, the highest dose levels evaluated, respectively. The corresponding plasma Cmax and AYClast for male and female rats were 214,000 and 229,000 ng/mL and 3,680,000 and 3,810,000 h * ng/mL, respectively. For male and female dogs, the corresponding plasma Cmax and AYClast were 76,000 and 92,400 ng/mL and 976,000 and 1,200,000 h * ng/mL, respectively.
Collapse
Affiliation(s)
- Joseph C. Cacaccio
- Photodynamic Therapy Center, Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (J.C.C.); (F.A.D.)
| | - Farukh A. Durrani
- Photodynamic Therapy Center, Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (J.C.C.); (F.A.D.)
| | | | - Ravindra K. Pandey
- Photodynamic Therapy Center, Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (J.C.C.); (F.A.D.)
| |
Collapse
|
29
|
Alphandéry E. Ultrasound and nanomaterial: an efficient pair to fight cancer. J Nanobiotechnology 2022; 20:139. [PMID: 35300712 PMCID: PMC8930287 DOI: 10.1186/s12951-022-01243-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/02/2022] [Indexed: 01/12/2023] Open
Abstract
Ultrasounds are often used in cancer treatment protocols, e.g. to collect tumor tissues in the right location using ultrasound-guided biopsy, to image the region of the tumor using more affordable and easier to use apparatus than MRI and CT, or to ablate tumor tissues using HIFU. The efficacy of these methods can be further improved by combining them with various nano-systems, thus enabling: (i) a better resolution of ultrasound imaging, allowing for example the visualization of angiogenic blood vessels, (ii) the specific tumor targeting of anti-tumor chemotherapeutic drugs or gases attached to or encapsulated in nano-systems and released in a controlled manner in the tumor under ultrasound application, (iii) tumor treatment at tumor site using more moderate heating temperatures than with HIFU. Furthermore, some nano-systems display adjustable sizes, i.e. nanobubbles can grow into micro-bubbles. Such dual size is advantageous since it enables gathering within the same unit the targeting properties of nano bubbles via EPR effect and the enhanced ultrasound contrasting properties of micro bubbles. Interestingly, the way in which nano-systems act against a tumor could in principle also be adjusted by accurately selecting the nano-system among a large choice and by tuning the values of the ultrasound parameters, which can lead, due to their mechanical nature, to specific effects such as cavitation that are usually not observed with purely electromagnetic waves and can potentially help destroying the tumor. This review highlights the clinical potential of these combined treatments that can improve the benefit/risk ratio of current cancer treatments.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS, 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de. Cosmochimie, IMPMC, 75005, Paris, France. .,Nanobacterie SARL, 36 boulevard Flandrin, 75116, Paris, France. .,Institute of Anatomy, UZH University of Zurich, Instiute of Anatomy, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| |
Collapse
|
30
|
Applications of Antimicrobial Photodynamic Therapy against Bacterial Biofilms. Int J Mol Sci 2022; 23:ijms23063209. [PMID: 35328629 PMCID: PMC8953781 DOI: 10.3390/ijms23063209] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 12/14/2022] Open
Abstract
Antimicrobial photodynamic therapy and allied photodynamic antimicrobial chemotherapy have shown remarkable activity against bacterial pathogens in both planktonic and biofilm forms. There has been little or no resistance development against antimicrobial photodynamic therapy. Furthermore, recent developments in therapies that involve antimicrobial photodynamic therapy in combination with photothermal hyperthermia therapy, magnetic hyperthermia therapy, antibiotic chemotherapy and cold atmospheric pressure plasma therapy have shown additive and synergistic enhancement of its efficacy. This paper reviews applications of antimicrobial photodynamic therapy and non-invasive combination therapies often used with it, including sonodynamic therapy and nanozyme enhanced photodynamic therapy. The antimicrobial and antibiofilm mechanisms are discussed. This review proposes that these technologies have a great potential to overcome the bacterial resistance associated with bacterial biofilm formation.
Collapse
|
31
|
Aghajanzadeh M, Zamani M, Rajabi Kouchi F, Eixenberger J, Shirini D, Estrada D, Shirini F. Synergic Antitumor Effect of Photodynamic Therapy and Chemotherapy Mediated by Nano Drug Delivery Systems. Pharmaceutics 2022; 14:pharmaceutics14020322. [PMID: 35214054 PMCID: PMC8880656 DOI: 10.3390/pharmaceutics14020322] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
This review provides a summary of recent progress in the development of different nano-platforms for the efficient synergistic effect between photodynamic therapy and chemotherapy. In particular, this review focuses on various methods in which photosensitizers and chemotherapeutic agents are co-delivered to the targeted tumor site. In many cases, the photosensitizers act as drug carriers, but this review, also covers different types of appropriate nanocarriers that aid in the delivery of photosensitizers to the tumor site. These nanocarriers include transition metal, silica and graphene-based materials, liposomes, dendrimers, polymers, metal–organic frameworks, nano emulsions, and biologically derived nanocarriers. Many studies have demonstrated various benefits from using these nanocarriers including enhanced water solubility, stability, longer circulation times, and higher accumulation of therapeutic agents/photosensitizers at tumor sites. This review also describes novel approaches from different research groups that utilize various targeting strategies to increase treatment efficacy through simultaneous photodynamic therapy and chemotherapy.
Collapse
Affiliation(s)
- Mozhgan Aghajanzadeh
- Department of Chemistry, College of Science, University of Guilan, Rasht 41335-19141, Iran; (M.A.); (M.Z.)
| | - Mostafa Zamani
- Department of Chemistry, College of Science, University of Guilan, Rasht 41335-19141, Iran; (M.A.); (M.Z.)
| | - Fereshteh Rajabi Kouchi
- Micron School of Materials Science and Engineering, Boise State University, Boise, ID 83725, USA; (F.R.K.); (D.E.)
| | - Josh Eixenberger
- Micron School of Materials Science and Engineering, Boise State University, Boise, ID 83725, USA; (F.R.K.); (D.E.)
- Center for Advanced Energy Studies, Boise State University, Boise, ID 83725, USA
- Correspondence: (J.E.); or (F.S.)
| | - Dorsa Shirini
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran;
| | - David Estrada
- Micron School of Materials Science and Engineering, Boise State University, Boise, ID 83725, USA; (F.R.K.); (D.E.)
- Center for Advanced Energy Studies, Boise State University, Boise, ID 83725, USA
| | - Farhad Shirini
- Department of Chemistry, College of Science, University of Guilan, Rasht 41335-19141, Iran; (M.A.); (M.Z.)
- Correspondence: (J.E.); or (F.S.)
| |
Collapse
|
32
|
Combinatorial Therapeutic Approaches with Nanomaterial-Based Photodynamic Cancer Therapy. Pharmaceutics 2022; 14:pharmaceutics14010120. [PMID: 35057015 PMCID: PMC8780767 DOI: 10.3390/pharmaceutics14010120] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/11/2021] [Accepted: 12/28/2021] [Indexed: 12/27/2022] Open
Abstract
Photodynamic therapy (PDT), in which a light source is used in combination with a photosensitizer to induce local cell death, has shown great promise in therapeutically targeting primary tumors with negligible toxicity and minimal invasiveness. However, numerous studies have shown that noninvasive PDT alone is not sufficient to completely ablate tumors in deep tissues, due to its inherent shortcomings. Therefore, depending on the characteristics and type of tumor, PDT can be combined with surgery, radiotherapy, immunomodulators, chemotherapy, and/or targeted therapy, preferably in a patient-tailored manner. Nanoparticles are attractive delivery vehicles that can overcome the shortcomings of traditional photosensitizers, as well as enable the codelivery of multiple therapeutic drugs in a spatiotemporally controlled manner. Nanotechnology-based combination strategies have provided inspiration to improve the anticancer effects of PDT. Here, we briefly introduce the mechanism of PDT and summarize the photosensitizers that have been tested preclinically for various cancer types and clinically approved for cancer treatment. Moreover, we discuss the current challenges facing the combination of PDT and multiple cancer treatment options, and we highlight the opportunities of nanoparticle-based PDT in cancer therapies.
Collapse
|
33
|
Luo L, Qi Y, Zhong H, Jiang S, Zhang H, Cai H, Wu Y, Gu Z, Gong Q, Luo K. GSH-sensitive polymeric prodrug: Synthesis and loading with photosensitizers as nanoscale chemo-photodynamic anti-cancer nanomedicine. Acta Pharm Sin B 2022; 12:424-436. [PMID: 35127396 PMCID: PMC8799999 DOI: 10.1016/j.apsb.2021.05.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/23/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022] Open
Abstract
Precisely delivering combinational therapeutic agents has become a crucial challenge for anti-tumor treatment. In this study, a novel redox-responsive polymeric prodrug (molecular weight, MW: 93.5 kDa) was produced by reversible addition-fragmentation chain transfer (RAFT) polymerization. The amphiphilic block polymer-doxorubicin (DOX) prodrug was employed to deliver a hydrophobic photosensitizer (PS), chlorin e6 (Ce6), and the as-prepared nanoscale system [NPs(Ce6)] was investigated as a chemo-photodynamic anti-cancer agent. The glutathione (GSH)-cleavable disulfide bond was inserted into the backbone of the polymer for biodegradation inside tumor cells, and DOX conjugated onto the polymer with a disulfide bond was successfully released intracellularly. NPs(Ce6) released DOX and Ce6 with their original molecular structures and degraded into segments with low MWs of 41.2 kDa in the presence of GSH. NPs(Ce6) showed a chemo-photodynamic therapeutic effect to kill 4T1 murine breast cancer cells, which was confirmed from a collapsed cell morphology, a lifted level in the intracellular reactive oxygen species, a reduced viability and induced apoptosis. Moreover, ex vivo fluorescence images indicated that NPs(Ce6) retained in the tumor, and exhibited a remarkable in vivo anticancer efficacy. The combinational therapy showed a significantly increased tumor growth inhibition (TGI, 58.53%). Therefore, the redox-responsive, amphiphilic block polymeric prodrug could have a great potential as a chemo-photodynamic anti-cancer agent.
Collapse
Affiliation(s)
- Lei Luo
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Yiming Qi
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Hong Zhong
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Shinan Jiang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA 91711, USA
| | - Hao Cai
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yahui Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Corresponding author. Tel./fax: +86 28 85422538.
| |
Collapse
|
34
|
Tu L, Liao Z, Luo Z, Wu Y, Herrmann A, Huo S. Ultrasound-controlled drug release and drug activation for cancer therapy. EXPLORATION (BEIJING, CHINA) 2021; 1:20210023. [PMID: 37323693 PMCID: PMC10190934 DOI: 10.1002/exp.20210023] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/02/2021] [Indexed: 06/15/2023]
Abstract
Traditional chemotherapy suffers from severe toxicity and side effects that limit its maximum application in cancer therapy. To overcome this challenge, an ideal treatment strategy would be to selectively control the release or regulate the activity of drugs to minimize the undesirable toxicity. Recently, ultrasound (US)-responsive drug delivery systems (DDSs) have attracted significant attention due to the non-invasiveness, high tissue penetration depth, and spatiotemporal controllability of US. Moreover, the US-induced mechanical force has been proven to be a robust method to site-selectively rearrange or cleave bonds in mechanochemistry. This review describes the US-activated DDSs from the fundamental basics and aims to present a comprehensive summary of the current understanding of US-responsive DDSs for controlled drug release and drug activation. First, we summarize the typical mechanisms for US-responsive drug release and drug activation. Second, the main factors affecting the ultrasonic responsiveness of drug carriers are outlined. Furthermore, representative examples of US-controlled drug release and drug activation are discussed, emphasizing their novelty and design principles. Finally, the challenges and an outlook on this promising therapeutic strategy are discussed.
Collapse
Affiliation(s)
- Li Tu
- Fujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenP. R. China
| | - Zhihuan Liao
- Fujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenP. R. China
| | - Zheng Luo
- Fujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenP. R. China
| | - Yun‐Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenP. R. China
| | - Andreas Herrmann
- DWI – Leibniz Institute for Interactive MaterialsAachenGermany
- Institute of Technical and Macromolecular ChemistryRWTH Aachen UniversityAachenGermany
| | - Shuaidong Huo
- Fujian Provincial Key Laboratory of Innovative Drug Target ResearchSchool of Pharmaceutical SciencesXiamen UniversityXiamenP. R. China
| |
Collapse
|
35
|
Li Y, Yu P, Fu W, Cai L, Yu Y, Feng Z, Wang Y, Zhang F, Yu X, Xu H, Sui D. Ginseng-Astragalus-oxymatrine injection ameliorates cyclophosphamide-induced immunosuppression in mice and enhances the immune activity of RAW264.7 cells. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114387. [PMID: 34216728 DOI: 10.1016/j.jep.2021.114387] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/23/2021] [Accepted: 06/30/2021] [Indexed: 05/14/2023]
Abstract
ETHNOPHARMACOLOGICAL SIGNIFICANCE Ginseng quinquefolium (L.), Astragalus membranaceus, and Sophora flavescens Aiton are popular folk medicines in many Asian countries and regions. These three traditional Chinese herbs and their extracts have been reported to considerably enhance the immune function. G. quinquefolium (L.) is considered the king of herbs in China. Traditionally, G. quinquefolium (L.) is believed to replenish vitality, which is considered as immune enhancement in modern Chinese pharmacy. One of the main uses of Astragalus is immunity enhancement; S. flavescens and oxymatrine obtained from its extract have been used to treat leukopenia. Considering the pharmacological properties of Ginseng, Astragalus, and oxymatrine, we evaluated the immunopotentiation effects of their combination, Ginseng-Astragalus-oxymatrine (GAO), in the present study. AIM OF THE STUDY This study aimed to expand the clinical application of GAO and to preliminarily explore its mechanism of action by determining whether GAO injection can enhance immunity in vivo and in vitro. METHODS Overall, 17 major chemical components in GAO were analysed using HPLC and LC-MS. The immunity-enhancing effect of GAO was studied in the cyclophosphamide (CTX)-induced immunosuppressive mouse model and RAW 264.7 cells. RESULTS Quantitative analysis showed that the potential active components of GAO include at least ginsenosides, astragaloside IV, and oxymatrine. GAO could significantly improve the nonspecific immunity including the indices of the thymus and spleen, number of peripheral blood leukocytes, levels of TNF-α and IL-6, phagocytic function of macrophages, and cytotoxic activity of natural killer (NK) cells. Additionally, GAO enhanced the humoural immunity, characterised by the antibody production ability of B cells, and cellular immunity, characterised by the activity of T cells, in immunosuppressed mouse. Moreover, GAO could enhance the phagocytic and adhesion functions of RAW 264.7 cells, which may be related to the activation of reactive oxygen species and NF-κB signalling pathway. CONCLUSION GAO could dramatically ameliorate CTX-induced immunosuppression in mouse and stimulate the immune activity in RAW 264.7 cells possibly by activating the NF-κB signalling pathway.
Collapse
Affiliation(s)
- Yuangeng Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China.
| | - Ping Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China.
| | - Wenwen Fu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China.
| | - Lijian Cai
- Changbaishan Pharmaceutical CO. LTD, Jiaohe, China.
| | - Ying Yu
- Changbaishan Pharmaceutical CO. LTD, Jiaohe, China.
| | | | - Yaozhen Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China.
| | - Fuyuan Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China.
| | - Xiaofeng Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China.
| | - Huali Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China.
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
36
|
Aziz B, Khurshid A, Mahmood R, Khan JA, Javaid S, Alam M, Mujtaba Ul Hassan S, Ikram M. Study of synergistic effects of Ficus Carica leaves extract mediated chemo-photodynamic therapy on rhabdomyosarcoma cells. Photodiagnosis Photodyn Ther 2021; 36:102565. [PMID: 34614426 DOI: 10.1016/j.pdpdt.2021.102565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Chemotherapy for rhabdomyosarcoma (RD) is effective, but it has critical side effects and unavoidable challenges. Photodynamic therapy (PDT) is an approach to treating cancer with relatively moderate side effects. Plant products are a rich source of polyphenols, which have potent antioxidant and anticancer activities. Therefore, their research has become an emerging field in recent decades. PURPOSE This work aimed to evaluate the potential of hydrophobic extract of Ficus Carica (FC) to determine whether FC in the presence of low dose chemo and Aluminium Phthalocyanine (Photosense®) mediated photodynamic therapy synergistically enhances the treatment efficacy of RD cells. METHOD FC with and without combination with individual therapeutic modalities like photosense mediated photodynamic therapy, chemotherapy, and their combinations were studied for cell viability and morphological changes in invitro RD cells. A semiconductor diode laser (630 nm) was used as a light source in PDT. The cytotoxic effect of FC on cell viability and cellular morphological changes were investigated by MTT reagent and a camera attached to an inverted visible light microscope. The effect of FC, followed by di-combination with low dose chemo (doxorubicin-HCl, and dacarbazine), Photosense® mediated PDT and chemo-Photosense® mediated PDT (tri-combination) at 630 nm diode laser and 10 J/cm2 fluency were also investigated by MTT reagent. The combination index method is used to identify the synergistic effect of combination therapy by using CompuSyn software based on the Chou-Talalay method. RESULTS The dose-dependent effect of FC on cell viability and cellular morphological changes were observed in the RD cell line. It was found that the pre incubation of FC potentiated the anticancer effect as a neoadjuvant agent for doxorubicin-HCl and decarbazine based chemotherapy, Photosense® mediated PDT and chemo-PDT (tri-combination) with synergistic effect (CI<1). CONCLUSION These results suggest a possible thread that the low dose combination of the aforementioned therapeutic modalities in the presence of FC remarkably enhances the treatment efficacy of RD in comparison with a single-agent treatment modality. The proposed sequence of FC with chemo and PDT might present better therapeutic outcomes in RD therapies and may provide result for RD metastasis. FC may also be used in the application of phyto-PDT to cancer in the future.
Collapse
Affiliation(s)
- Bushra Aziz
- Photonanomedicine Research Laboratory, Department of Physics and Applied Mathematics, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nilore, Islamabad 45650, Pakistan; Department of Physics, Women University of Azad Jammu & Kashmir, Bagh, Azad Kashmir, Pakistan
| | - Ahmat Khurshid
- Photonanomedicine Research Laboratory, Department of Physics and Applied Mathematics, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nilore, Islamabad 45650, Pakistan.
| | - Rashid Mahmood
- Photonanomedicine Research Laboratory, Department of Physics and Applied Mathematics, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nilore, Islamabad 45650, Pakistan
| | - Junaid Ahmad Khan
- Photonanomedicine Research Laboratory, Department of Physics and Applied Mathematics, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nilore, Islamabad 45650, Pakistan
| | - Sumbal Javaid
- Photonanomedicine Research Laboratory, Department of Physics and Applied Mathematics, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nilore, Islamabad 45650, Pakistan; Department of Animal Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Masroor Alam
- Department of Virology and Imunology, National Institute of Health, Park Road, Islamabad, Pakistan
| | - Syed Mujtaba Ul Hassan
- Department of Metallurgy and Materials Engineering, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nilore, Islamabad 45650, Pakistan
| | - Masroor Ikram
- Photonanomedicine Research Laboratory, Department of Physics and Applied Mathematics, Pakistan Institute of Engineering and Applied Sciences (PIEAS), Nilore, Islamabad 45650, Pakistan
| |
Collapse
|
37
|
Li G, Li J, Hou Y, Xie S, Xu J, Yang M, Li D, Du Y. Levofloxacin-Loaded Nanosonosensitizer as a Highly Efficient Therapy for Bacillus Calmette-Guérin Infections Based on Bacteria-Specific Labeling and Sonotheranostic Strategy. Int J Nanomedicine 2021; 16:6553-6573. [PMID: 34602818 PMCID: PMC8478796 DOI: 10.2147/ijn.s321631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose The rapid emergence of multidrug-resistant Mycobacterium tuberculosis (MTB) poses a significant challenge to the treatment of tuberculosis (TB). Sonodynamic antibacterial chemotherapy (SACT) combined with sonosensitizer-loaded nanoparticles with targeted therapeutic function is highly expected to eliminate bacteria without fear of drug resistance. This study aimed to investigate the antibacterial effect and underlying mechanism of levofloxacin-loaded nanosonosensitizer with targeted therapeutic function against Bacillus Calmette-Guérin bacteria (BCG, an MTB model). Methods This study developed levofloxacin-loaded PLGA-PEG (poly lactide-co-glycolide-polyethylene glycol) nanoparticles with BM2 aptamer conjugation on its surface using the crosslinking agents EDC and NHS (BM2-LVFX-NPs). The average diameter, zeta potential, morphology, drug-loading properties, and drug release efficiency of the BM2-LVFX-NPs were investigated. In addition, the targeting and toxicity of BM2-LVFX-NPs in the subcutaneous BCG infection model were evaluated. The biosafety, reactive oxygen species (ROS) production, cellular phagocytic effect, and antibacterial effect of BM2-LVFX-NPs in the presence of ultrasound stimulations (42 kHz, 0.67 W/cm2, 5 min) were also systematically evaluated. Results BM2-LVFX-NPs not only specifically recognized BCG bacteria in vitro but also gathered accurately in the lesion tissues. Drugs loaded in BM2-LVFX-NPs with the ultrasound-responsive feature were effectively released compared to the natural state. In addition, BM2-LVFX-NPs exhibited significant SACT efficiency with higher ROS production levels than others, resulting in the effective elimination of bacteria in vitro. Meanwhile, in vivo experiments, compared with other options, BM2-LVFX-NPs also exhibited an excellent therapeutic effect in a rat model with BCG infection after exposure to ultrasound. Conclusion Our work demonstrated that a nanosonosensitizer formulation with LVFX could efficiently translocate therapeutic drugs into the cell and improve the bactericidal effects under ultrasound, which could be a promising strategy for targeted therapy for MTB infections with high biosafety.
Collapse
Affiliation(s)
- Gangjing Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jianhu Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yuru Hou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Shuang Xie
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jieru Xu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Min Yang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Dairong Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yonghong Du
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China.,Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| |
Collapse
|
38
|
Cancer-cell-biomimetic Upconversion nanoparticles combining chemo-photodynamic therapy and CD73 blockade for metastatic triple-negative breast cancer. J Control Release 2021; 337:90-104. [PMID: 34274385 DOI: 10.1016/j.jconrel.2021.07.021] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/28/2021] [Accepted: 07/13/2021] [Indexed: 12/15/2022]
Abstract
Photodynamic therapy (PDT) and chemotherapy show clinical promise in destroying orthotopic tumors but are insufficient against abscopal metastases. The research reports the combined application of an anti-CD73 antibody and chemo-PDT to synergistically amplify the anti-metastatic effects of T cell-mediated antitumor immunity. The cancer cell membrane (CM)-cloaked upconversion nanoparticles, integrating rose bengal (RB) and the reactive oxygen species (ROS)-sensitive polymer polyethylene glycol-thioketal-doxorubicin (PEG-TK-DOX, i.e., PTD), are tailored for near-infrared (NIR)-triggered chemo-PDT. CM camouflage enables nanoparticles' excellent tumor-targeting abilities and immune escape from macrophages. The combination of PDT and chemotherapy presents strong synergistic antitumor efficacy and synchronously causes a series of immunogenic cell death (ICD), leading to tumor-specific immunity. The anti-CD73 antibody prevents the immunosuppression phenomenon in tumors by blocking the adenosine pathway, and it is emerging as a sufficient immune checkpoint blockade when combined with ICD-elicited tumor therapies. As cancer membrane camouflaged nanoparticles CM@UCNP-RB/PTD combined with anti-CD73 antibodies, synergistic efficacy of chemotherapy and PDT not only destroys the orthotopic tumors by DOX and cytotoxic ROS but also prevents abscopal tumor metastasis via inducing systemic cytotoxic T cell responses with CD73 blockade. This strategy is promising in curing metastatic triple-negative breast cancer in preclinical research.
Collapse
|
39
|
El-Hussein A, Manoto SL, Ombinda-Lemboumba S, Alrowaili ZA, Mthunzi-Kufa P. A Review of Chemotherapy and Photodynamic Therapy for Lung Cancer Treatment. Anticancer Agents Med Chem 2021; 21:149-161. [PMID: 32242788 DOI: 10.2174/1871520620666200403144945] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/15/2020] [Accepted: 02/20/2020] [Indexed: 11/22/2022]
Abstract
Cancer is among the leading causes of mortality and morbidity worldwide. Among the different types of cancers, lung cancer is considered to be the leading cause of death related to cancer and the most commonly diagnosed form of such disease. Chemotherapy remains a dominant treatment modality for many types of cancers at different stages. However, in many cases, cancer cells develop drug resistance and become nonresponsive to chemotherapy, thus, necessitating the exploration of alternative and /or complementary treatment modalities. Photodynamic Therapy (PDT) has emerged as an effective treatment modality for various malignant neoplasia and tumors. In PDT, the photochemical interaction of light, Photosensitizer (PS) and molecular oxygen produces Reactive Oxygen Species (ROS), which induces cell death. Combination therapy, by using PDT and chemotherapy, can promote synergistic effect against this fatal disease with the elimination of drug resistance, and enhancement of the efficacy of cancer eradication. In this review, we give an overview of chemotherapeutic modalities, PDT, and the different types of drugs associated with each therapy. Furthermore, we also explored the combined use of chemotherapy and PDT in the course of lung cancer treatment and how this approach could be the last resort for thousands of patients that have been diagnosed by this fatal disease.
Collapse
Affiliation(s)
- Ahmed El-Hussein
- National Institute of Laser Enhanced Science, Cairo University, Giza, Egypt
| | - Sello L Manoto
- Council for Scientific and Industrial Research (CSIR), National Laser Centre, Pretoria, South Africa
| | | | - Ziya A Alrowaili
- Physics Department, College of Science, Jouf University, Jouf, Saudi Arabia
| | - Patience Mthunzi-Kufa
- Council for Scientific and Industrial Research (CSIR), National Laser Centre, Pretoria, South Africa
| |
Collapse
|
40
|
Wang F, Chen J, Liu J, Zeng H. Cancer theranostic platforms based on injectable polymer hydrogels. Biomater Sci 2021; 9:3543-3575. [PMID: 33634800 DOI: 10.1039/d0bm02149k] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Theranostic platforms that combine therapy with diagnosis not only prevent the undesirable biological responses that may occur when these processes are conducted separately, but also allow individualized therapies for patients. Polymer hydrogels have been employed to provide well-controlled drug release and targeted therapy in theranostics, where injectable hydrogels enable non-invasive treatment and monitoring with a single injection, offering greater patient comfort and efficient therapy. Efforts have been focused on applying injectable polymer hydrogels in theranostic research and clinical use. This review highlights recent progress in the design of injectable polymer hydrogels for cancer theranostics, particularly focusing on the elements/components of theranostic hydrogels, and their cross-linking strategies, structures, and performance with regard to drug delivery/tracking. Therapeutic agents and tracking modalities that are essential components of the theranostic platforms are introduced, and the design strategies, properties and applications of the injectable hydrogels developed via two approaches, namely chemical bonds and physical interactions, are described. The theranostic functions of the platforms are highly dependent on the architecture and components employed for the construction of hydrogels. Challenges currently presented by theranostic platforms based on injectable hydrogels are identified, and prospects of acquiring more comfortable and personalized therapies are proposed.
Collapse
Affiliation(s)
- Feifei Wang
- The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510700, China. and Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada.
| | - Jingsi Chen
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada.
| | - Jifang Liu
- The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510700, China.
| | - Hongbo Zeng
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada.
| |
Collapse
|
41
|
Yang L, He X, Zeng Z, Tang J, Qi D, Ma H, Chen H, Ning X, Feng X. Clickable amino acid tuned self-assembly of a nucleus-selective multi-component nanoplatform for synergistic cancer therapy. Chem Sci 2021; 12:8394-8400. [PMID: 34221320 PMCID: PMC8221198 DOI: 10.1039/d1sc01073e] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/13/2021] [Indexed: 11/21/2022] Open
Abstract
Nucleus-targeted therapy holds great promise in cancer treatment; however, a lack of effective nucleus-specific delivery significantly limits its application potential. Here, we report a nucleus-targeted synergistic chemo-photodynamic therapy based on the self-assembly of chlorin e6 (Ce6) and doxorubicin (DOX) tuned by clickable dibenzocyclooctyne (DIBO) functionalized lysine (D-K) and subsequent reaction with crosslinkers. The assembled nanodrugs with high loading efficiency and long-term stability show enhanced cellular uptake and accumulation in the nucleus, resulting in greatly improved in vitro and in vivo chemo-photodynamic efficacy. Notably, D-K can promote the rapid self-assembly of Ce6 and DOX in aqueous solution, avoiding the introduction of organic solvents or tedious preparations. In addition, the introduction of the DIBO group can effectively expand the types of self-assembly material and enhance the self-assembly behaviour through a copper-free click reaction. Therefore, we present an effective nucleus-targeted combination drug delivery strategy, which has great potential in the treatment of many diseases.
Collapse
Affiliation(s)
- Lan Yang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 China
| | - Xiao He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 China
| | - Zhiying Zeng
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 China
| | - Jiakun Tang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 China
| | - Dongmei Qi
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 China
| | - Huijie Ma
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 China
| | - Hui Chen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University Nanjing 210093 China
| | - Xuli Feng
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University Chongqing 401331 China
| |
Collapse
|
42
|
Zhang Y, Zhou T, Li J, Xu N, Cai M, Zhang H, Zhao Q, Wang S. Au Catalyzing Control Release NO in vivo and Tumor Growth-Inhibiting Effect in Chemo-Photothermal Combination Therapy. Int J Nanomedicine 2021; 16:2501-2513. [PMID: 33824588 PMCID: PMC8018432 DOI: 10.2147/ijn.s270466] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 02/27/2021] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Aim to obtain a NO donor that can control released NO in vivo with the high efficacy of tumor suppression and targeting, a nanoplatform consisting of FA-Fe3O4@mSiO2-Au/DOX was constructed. METHODS In vitro, the nanoplatform catalyzed NO's release with the maximum value of 4.91 μM within 60 min at 43°C pH=5.0, which was increased by 1.14 times when the temperature was 37°C. In vivo, 11.7 μg Au in the tumor tissue was found to catalyze S-nitrosoglutathione continuously, and 54 μM NO was checked out in the urine. RESULTS AND DISCUSSION The high concentration of NO was found to increase the apoptotic rate and to reduce tumor proliferation. In the chemo-photothermal combination therapy, the tumor inhibition rate was increased up to 94.3%, and Au's contribution from catalyzing NO release NO was 8.17%.
Collapse
Affiliation(s)
- Ying Zhang
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Tianfu Zhou
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Jian Li
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Nuo Xu
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Mingze Cai
- Key Laboratory of TargetDrug Design and Research, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Hong Zhang
- Van ’T Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, 1098 XH, the Netherlands
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| |
Collapse
|
43
|
Du C, Li S, Li Y, Galons H, Guo N, Teng Y, Zhang Y, Li M, Yu P. F7 and topotecan co-loaded thermosensitive liposome as a nano-drug delivery system for tumor hyperthermia. Drug Deliv 2021; 27:836-847. [PMID: 32508162 PMCID: PMC8216433 DOI: 10.1080/10717544.2020.1772409] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In order to enhance the targeting efficiency and reduce anti-tumor drug’s side effects, topotecan (TPT) and F7 were co-loaded in thermosensitive liposomes (F7-TPT-TSL), which show enhanced permeability and retention in tumors, as well as local controlled release by heating in vitro. TPT is a water-soluble inhibitor of topoisomerase I that is converted to an inactive carboxylate structure under physiological conditions (pH 7.4). F7 is a novel drug significantly resistant to cyclin-dependent kinase but its use was restricted by its high toxicity. F7-TPT-TSL had excellent particle distribution (about 103 nm), high entrapment efficiency (>95%), obvious thermosensitive property, and good stability. Confocal microscopy demonstrated specific higher accumulation of TSL in tumor cells. MTT proved F7-TPT-TSL/H had strongest cell lethality compared with other formulations. Then therapeutic efficacy revealed synergism of TPT and F7 co-loaded in TSL, together with hyperthermia. Therefore, the F7-TPT-TSL may serve as a promising system for temperature triggered cancer treatment.
Collapse
Affiliation(s)
- Chunyang Du
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Shuangshuang Li
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Yuan Li
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Hervé Galons
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China.,Institut Parisien de Chimie Moléculaire, UMR CNRS 8232, Paris, France
| | - Na Guo
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Yuou Teng
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Yongmin Zhang
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China.,Institut Parisien de Chimie Moléculaire, UMR CNRS 8232, Paris, France
| | - Mingyuan Li
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Peng Yu
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| |
Collapse
|
44
|
Zhong Y, Dong Y, Chen T, Yang L, Yao M, Zhi Y, Yang H, Zhang J, Bi W. 808 nm NIR Laser-Excited Upconversion Nanoplatform for Combinatory Photodynamic and Chemotherapy with Deep Penetration and Acid Bursting Release Performance. ACS APPLIED BIO MATERIALS 2021; 4:2639-2653. [DOI: 10.1021/acsabm.0c01607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Yingtao Zhong
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Yun Dong
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Tie Chen
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Lingzhi Yang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Min Yao
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Yunshi Zhi
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Haoyi Yang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Jian Zhang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| | - Wenchuan Bi
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
45
|
Zhang L, Jin T, Sun J, Chen X. Self-assembly supramolecular drug delivery system for combination of photodynamic therapy and chemotherapy. J Microencapsul 2021; 38:81-88. [PMID: 32964772 DOI: 10.1080/02652048.2020.1826591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/14/2020] [Indexed: 10/23/2022]
Abstract
AIMS To construct a self-assembly supramolecular drug delivery system (DDS) to co-deliver chlorin e6 (Ce6) and tripeptide tyroseroleutide (YSL) and evaluate the anti-tumour effects. METHODS A supramolecular DDS was constructed via self-assembly of Ce6 and YSL based on π-π stacking and hydrogen-bond interaction. The size, morphology, stability, in vitro drug release, cellular uptake, cytotoxicity, pharmacokinetics analysis and pharmacodynamics analysis were respectively studied. RESULTS Ce6-YSL nanoparticles with a uniform size of 75 ± 3.5 nm (PDI = 0.128) and monodispersed spherical morphology were constructed. The nanoparticles exhibited good stability with zeta potential -21.2 ± 1.73 mV. Under the weak acidic conditions, the accumulative drug release was 82.8% (w/w) (pH = 6.0) and 91.5% (w/w) (pH = 5.0), respectively, indicating that nanoparticles performed smart responsive properties and achieved controlled release characteristics in acidic tumour microenvironment. In addition, nanoparticles could easily enter the tumour cells and induce ROS production and inhibit cell proliferation in SMMCC-7721 cells with IC50 value 3.4 ± 0.023 μg/mL under laser irradiation. Furthermore, the nanoparticles could retain a much higher blood concentration in vivo and displayed excellent antitumor effect in tumour-bearing mice, showing no influence on body weight. CONCLUSIONS This self-assembly supramolecular DDS can be used for combination of photodynamic therapy and chemotherapy in future research.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Taiyu Jin
- Department of Pharmacy, The Second People's Hospital of Lianyungang, Lianyungang, China
| | - Jing Sun
- Department of Pharmacy, The Second Hospital of Shandong University, Jinan, China
| | - Xiaoyu Chen
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
46
|
Exosome-based photoacoustic imaging guided photodynamic and immunotherapy for the treatment of pancreatic cancer. J Control Release 2021; 330:293-304. [PMID: 33359580 DOI: 10.1016/j.jconrel.2020.12.039] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/15/2020] [Accepted: 12/20/2020] [Indexed: 01/09/2023]
Abstract
Exosomes, which are released from all cells and take part in cell-to-cell communication, have been utilized as drug delivery vehicles in many recent studies. Immunotherapy is an emerging technology which uses patients' innate immune systems. In immunotherapy, immune cells are stimulated through antibodies, the other immune cells and genetic modifications for the purposes of, for instance, cancer therapy. In this study, tumor-derived re-assembled exosome (R-Exo) was simultaneously utilized as both a drug delivery carrier and an immunostimulatory agent. A chlorin e6 photosensitizer was loaded into tumor-derived exosomes during exosomal re-assembly. After this modification, R-Exo retains its original average size and has the same membrane proteins, which allows for targeting of tumor cells. Chlorin e6-loaded R-Exo (Ce6-R-Exo) can be visualized by photoacoustic imaging and can efficiently generate reactive oxygen species inside tumor cells under laser irradiation. In addition, Ce6-R-Exo increased the release of cytokines from immune cells, which indicates that these modified exosomes can be used as an immunotherapeutic agent. In conclusion, we developed a novel strategy that enables photoacoustic imaging-guided photodynamic and immune-combination therapy for the treatment of cancer with tumor-derived Ce6-R-Exo.
Collapse
|
47
|
Dai J, Chen J, Qi J, Ding M, Liu W, Shao T, Han J, Wang G. Konjac Glucomannan from Amorphophallus konjac enhances immunocompetence of the cyclophosphamide-induced immunosuppressed mice. Food Sci Nutr 2021; 9:728-735. [PMID: 33598158 PMCID: PMC7866624 DOI: 10.1002/fsn3.2038] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/24/2020] [Accepted: 11/13/2020] [Indexed: 01/19/2023] Open
Abstract
This present study was designed to evaluate the immunomodulatory activity of Konjac glucomannan (KGM) on immunosuppressed mice induced by cyclophosphamide (CTX) treatment. The mice immunodeficiency model was established by CTX. KGM was used to modulate the activities of immunosuppressive mice. It was proved that KGM could promote the proliferation of lymphocyte, thymus, and spleen indices, and alleviate the atrophy of immune organs and weight loss. Besides, in mice serum, the levels of cytokines including TNF-α, IgG, IL-2, and the contents of hemolysin were also increased after treatment with KGM. Furtherly, in nonspecific immunity, KGM could enhance natural killer (NK) cell lethality and pinocytic activity of mouse peritoneal macrophages. Therefore, all of these results revealed that KGM could improve the reduced immunity of CTX-induced mice via modulation innate immunity and adaptive immunity.
Collapse
Affiliation(s)
- Jiajia Dai
- School of Public HealthWannan Medical CollegeWuhuChina
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Provincial Engineering Laboratory for Screening and Re‐evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Province Key Laboratory of Active Biological MacromoleculesWuhuChina
| | - Jian Chen
- School of Public HealthWannan Medical CollegeWuhuChina
| | - Jun Qi
- College of Tea & Food Science and TechnologyAnhui Engineering Laboratory for Agro‐products ProcessingAnhui Agricultural UniversityHefeiChina
| | - Mengru Ding
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Provincial Engineering Laboratory for Screening and Re‐evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Province Key Laboratory of Active Biological MacromoleculesWuhuChina
- School of Pharmacy, Drug Research & Development CenterWannan Medical CollegeWuhuChina
| | - Wei Liu
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Provincial Engineering Laboratory for Screening and Re‐evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Province Key Laboratory of Active Biological MacromoleculesWuhuChina
- School of Pharmacy, Drug Research & Development CenterWannan Medical CollegeWuhuChina
| | - Taili Shao
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Provincial Engineering Laboratory for Screening and Re‐evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Province Key Laboratory of Active Biological MacromoleculesWuhuChina
- School of Pharmacy, Drug Research & Development CenterWannan Medical CollegeWuhuChina
| | - Jun Han
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Provincial Engineering Laboratory for Screening and Re‐evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Province Key Laboratory of Active Biological MacromoleculesWuhuChina
- School of Pharmacy, Drug Research & Development CenterWannan Medical CollegeWuhuChina
| | - Guodong Wang
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Provincial Engineering Laboratory for Screening and Re‐evaluation of Active Compounds of Herbal Medicines in Southern Anhui, Anhui Province Key Laboratory of Active Biological MacromoleculesWuhuChina
- School of Pharmacy, Drug Research & Development CenterWannan Medical CollegeWuhuChina
| |
Collapse
|
48
|
Zhang H, Wu Y, Xu X, Chen C, Xue X, Xu B, Li T, Chen Z. Synthesis Characterization of Platinum (IV) Complex Curcumin Backboned Polyprodrugs: In Vitro Drug Release Anticancer Activity. Polymers (Basel) 2020; 13:E67. [PMID: 33375302 PMCID: PMC7795977 DOI: 10.3390/polym13010067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
The conventional mono-chemotherapy still suffers from unsatisfied potency for cancer therapy due to tumor heterogeneity and the occurrence of drug resistance. Combination chemotherapy based on the nanosized drug delivery systems (nDDSs) has been developed as a promising platform to circumvent the limitations of mono-chemotherapy. In this work, starting from cisplatin and curcumin (Cur), we prepared a dual drug backboned shattering polymeric nDDS for synergistic chemotherapy. By in situ polymerization of the Cur, platinum (IV) complex-based prodrug monomer (DHP), L-lysine diisocyanate (LDI), and then conjugation with a hydrophilic poly (ethylene glycol) monomethyl ether (mPEG) derivative, a backbone-type platinum (IV) and Cur linkage containing mPEG-poly(platinum-co-Cur)-mPEG (PCPt) copolymer was synthesized. Notably, the platinum (IV) (Pt (IV)) and Cur were incorporated into the hydrophobic segment of PCPt with the fixed drugs loading ratio and high drugs loading content. The batch-to-batch variability could be decreased. The resulting prodrug copolymer then self-assembled into nanoparticles (PCPt NPs) with an average diameter around 100 nm, to formulate a synergetic nDDS. Importantly, PCPt NPs could greatly improve the solubility and stability of Cur. In vitro drug release profiles have demonstrated that PCPt NPs were stable in PBS 7.4, rapid burst release was greatly decreased, and the Pt and Cur release could be largely enhanced under reductive conditions due to the complete dissociation of the hydrophobic main chain of PCPt. In vitro cell viability test indicated that PCPt NPs were efficient synergistic chemotherapy units. Moreover, PCPt NPs were synergistic for cisplatin-resistant cell lines A549/DDP cells, and they exhibited excellent reversal ability of tumor resistance to cisplatin. This work provides a promising strategy for the design and synthesis of nDDS for combination chemotherapy.
Collapse
Affiliation(s)
- Honglei Zhang
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (H.Z.); (X.X.); (B.X.); (T.L.)
| | - Yanjuan Wu
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (H.Z.); (X.X.); (B.X.); (T.L.)
| | - Xiao Xu
- Institute of Food Safety and Environment Monitoring, College of Chemistry, Fuzhou University, Fuzhou 350108, China; (X.X.); (C.C.)
| | - Chen Chen
- Institute of Food Safety and Environment Monitoring, College of Chemistry, Fuzhou University, Fuzhou 350108, China; (X.X.); (C.C.)
| | - Xiukun Xue
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (H.Z.); (X.X.); (B.X.); (T.L.)
| | - Ben Xu
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (H.Z.); (X.X.); (B.X.); (T.L.)
| | - Tianduo Li
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (H.Z.); (X.X.); (B.X.); (T.L.)
| | - Zhaowei Chen
- Institute of Food Safety and Environment Monitoring, College of Chemistry, Fuzhou University, Fuzhou 350108, China; (X.X.); (C.C.)
| |
Collapse
|
49
|
Verebová V, Beneš J, Staničová J. Biophysical Characterization and Anticancer Activities of Photosensitive Phytoanthraquinones Represented by Hypericin and Its Model Compounds. Molecules 2020; 25:E5666. [PMID: 33271809 PMCID: PMC7731333 DOI: 10.3390/molecules25235666] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 12/17/2022] Open
Abstract
Photosensitive compounds found in herbs have been reported in recent years as having a variety of interesting medicinal and biological activities. In this review, we focus on photosensitizers such as hypericin and its model compounds emodin, quinizarin, and danthron, which have antiviral, antifungal, antineoplastic, and antitumor effects. They can be utilized as potential agents in photodynamic therapy, especially in photodynamic therapy (PDT) for cancer. We aimed to give a comprehensive summary of the physical and chemical properties of these interesting molecules, emphasizing their mechanism of action in relation to their different interactions with biomacromolecules, specifically with DNA.
Collapse
Affiliation(s)
- Valéria Verebová
- Department of Chemistry, Biochemistry and Biophysics, University of Veterinary Medicine & Pharmacy, Komenského 73, 041 81 Košice, Slovakia;
| | - Jiří Beneš
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Kateřinská 1, 121 08 Prague, Czech Republic;
| | - Jana Staničová
- Department of Chemistry, Biochemistry and Biophysics, University of Veterinary Medicine & Pharmacy, Komenského 73, 041 81 Košice, Slovakia;
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Kateřinská 1, 121 08 Prague, Czech Republic;
| |
Collapse
|
50
|
Najafi M, Mortezaee K, Rahimifard M, Farhood B, Haghi-Aminjan H. The role of curcumin/curcuminoids during gastric cancer chemotherapy: A systematic review of non-clinical study. Life Sci 2020; 257:118051. [DOI: 10.1016/j.lfs.2020.118051] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023]
|