1
|
Pal M, Bera A, Masarkar N, Upadhyay A, Mukherjee S, Roy M. Targeted Chemo-Phototherapy in Red Light with Novel Doxorubicin and Iron(III) Complex-Functionalized Gold Nanoconjugate (Dox-Fe@FA-AuNPs). Chem Asian J 2024; 19:e202400616. [PMID: 38923831 DOI: 10.1002/asia.202400616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024]
Abstract
The anticancer efficacy of doxorubicin, an anthracycline-based and FDA-approved chemotherapeutic drug, is significantly hindered by acquired chemoresistance and severe side effects despite its potent anticancer properties. To overcome these challenges, we developed an innovative therapeutic formulation that integrates targeted chemotherapy and phototherapy within a single platform using gold nanoparticles (AuNPs). This novel nanoconjugate, designated as Dox-Fe@FA-AuNPs, is co-functionalized with folic acid, doxorubicin, and an iron(III)-phenolate/carboxylate complex, enabling cancer-specific drug activation. Here, we report the synthesis, characterization, and comprehensive physico-chemical and biological evaluations of Dox-Fe@FA-AuNPs. The nanoconjugate exhibited excellent solubility, stability, and enhanced cellular uptake in folate receptor-positive cancer cells. The nanoconjugate was potently cytotoxic against HeLa and MDA-MB-231 cancer cells (HeLa: 105.5±16.52 μg mL-1; MDA-MB-231: 112.0±12.31 μg mL-1; MDA-MB-231 (3D): 156.31±19.35 μg mL-1) while less cytotoxic to the folate(-) cancer cells (MCF-7, A549 and HepG2). The cytotoxicity was attributed to the pH-dependent release of doxorubicin, which preferentially occurs in the acidic tumor microenvironment. Additionally, under red light irradiation, the nanoconjugate generated ROS, inducing caspase-3/7-dependent apoptosis with a photo-index (PI) >50, and inhibited cancer cell migration. Our findings underscore the potential of Dox-Fe@FA-AuNPs as a highly effective and sustainable platform for targeted chemo-phototherapy.
Collapse
Affiliation(s)
- Maynak Pal
- Department of Chemistry, National Institute of Technology Manipur, Langol, 795004, Imphal West, Manipur
| | - Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore, Bangalore, 560012, Karnataka
| | - Neha Masarkar
- Department of Biochemistry, AIIMS Bhopal, Saket Nagar, Bhopal, Madhya Pradesh
| | - Aarti Upadhyay
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore, Bangalore, 560012, Karnataka
| | - Sukhes Mukherjee
- Department of Biochemistry, AIIMS Bhopal, Saket Nagar, Bhopal, Madhya Pradesh
| | - Mithun Roy
- Department of Chemistry, National Institute of Technology Manipur, Langol, 795004, Imphal West, Manipur
- Department of Chemistry, National Institute of Technology Agartala, Jirania, 799046, Tripura West
| |
Collapse
|
2
|
Zhang B, Man J, Guo L, Ru X, Zhang C, Liu W, Li L, Ma S, Guo L, Wang H, Wang B, Diao H, Che R, Yan L. Layer-by-Layer Nanoparticles for Calcium Overload in situ Enhanced Reactive Oxygen Oncotherapy. Int J Nanomedicine 2024; 19:7307-7321. [PMID: 39050879 PMCID: PMC11268784 DOI: 10.2147/ijn.s464981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
Background Challenges such as poor drug selectivity, non-target reactivity, and the development of drug resistance continue to pose significant obstacles in the clinical application of cancer therapeutic drugs. To overcome the limitations of drug resistance in chemotherapy, a viable treatment strategy involves designing multifunctional nano-platforms that exploit the unique physicochemical properties of tumor microenvironment (TME). Methods Herein, layer-by-layer nanoparticles with polyporous CuS as delivery vehicles, loaded with a sonosensitizer (tetra-(4-aminophenyl) porphyrin, TAPP) and sequentially functionalized with pH-responsive CaCO3, targeting group hyaluronic acid (HA) were designed and synthesized for synergistic treatment involving chemodynamic therapy (CDT), sonodynamic therapy (SDT), photothermal therapy (PTT), and calcium overload. Upon cleavage in an acidic environment, CaCO3 nanoparticles released TAPP and Ca2+, with TAPP generating 1O2 under ultrasound trigger. Exposed CuS produced highly cytotoxic ·OH in response to H2O2 and also exhibited a strong PTT effect. Results CuS@TAPP-CaCO3/HA (CTCH) delivered an enhanced ability to release more Ca2+ under acidic conditions with a pH value of 6.5, which in situ causes damage to HeLa mitochondria. In vitro and in vivo experiments both demonstrated that mitochondrial dysfunction greatly amplified the damage caused by reactive oxygen species (ROS) to tumor, which strongly confirms the synergistic effect between calcium overload and reactive oxygen therapy. Conclusion Collectively, the development of CTCH presents a novel therapeutic strategy for tumor treatment by effectively responding to the acidic TME, thus holding significant clinical implications.
Collapse
Affiliation(s)
- Boye Zhang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
- Shanxi Province Brain Degenerative Diseases Precision Diagnosis and Treatment Engineering Research Center, Shanxi Medical University, Jinzhong, 030606, People’s Republic of China
| | - Jianliang Man
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
- College of Pharmacy, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
| | - Lingyun Guo
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
| | - Xiaoxia Ru
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
| | - Chengwu Zhang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
- Shanxi Province Brain Degenerative Diseases Precision Diagnosis and Treatment Engineering Research Center, Shanxi Medical University, Jinzhong, 030606, People’s Republic of China
| | - Wen Liu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
- Shanxi Province Brain Degenerative Diseases Precision Diagnosis and Treatment Engineering Research Center, Shanxi Medical University, Jinzhong, 030606, People’s Republic of China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
| | - Lihong Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
- Shanxi Province Brain Degenerative Diseases Precision Diagnosis and Treatment Engineering Research Center, Shanxi Medical University, Jinzhong, 030606, People’s Republic of China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
| | - Sufang Ma
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
- Shanxi Province Brain Degenerative Diseases Precision Diagnosis and Treatment Engineering Research Center, Shanxi Medical University, Jinzhong, 030606, People’s Republic of China
| | - Lixia Guo
- Shanxi Province Brain Degenerative Diseases Precision Diagnosis and Treatment Engineering Research Center, Shanxi Medical University, Jinzhong, 030606, People’s Republic of China
- College of Pharmacy, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
| | - Haojiang Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
- Shanxi Province Brain Degenerative Diseases Precision Diagnosis and Treatment Engineering Research Center, Shanxi Medical University, Jinzhong, 030606, People’s Republic of China
| | - Bin Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
- Shanxi Province Brain Degenerative Diseases Precision Diagnosis and Treatment Engineering Research Center, Shanxi Medical University, Jinzhong, 030606, People’s Republic of China
| | - Haipeng Diao
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
| | - Renchao Che
- Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Academy for Engineering &Technology, Fudan University, Shanghai, 200438, People’s Republic of China
- Zhejiang Laboratory, Hangzhou, 311100, People’s Republic of China
| | - Lili Yan
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
- Shanxi Province Brain Degenerative Diseases Precision Diagnosis and Treatment Engineering Research Center, Shanxi Medical University, Jinzhong, 030606, People’s Republic of China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, People’s Republic of China
| |
Collapse
|
3
|
Qiao Y, Tang X, Qiuju X, Zhang G. Enzyme-loaded manganese-porphyrin metal-organic nanoframeworks for oxygen-evolving photodynamic therapy of hypoxic cells. Heliyon 2024; 10:e33902. [PMID: 39071555 PMCID: PMC11282992 DOI: 10.1016/j.heliyon.2024.e33902] [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: 03/25/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/30/2024] Open
Abstract
Photodynamic therapy (PDT) is attracting great attention for cancer treatments, while its therapeutic efficacy is limited by unsatisfactory photosensitizers and hypoxic tumor microenvironment (TME). To address these problems, we have developed catalase-loaded manganese-porphyrin frameworks (CAT@MnPFs) for catalytically-assisted PDT of cancer cells. CAT@MnPFs were constructed by the assembly of Mn2+ ions and PpIX into MnPFs and the subsequent loading of catalase. Under 650 nm light irradiation, the porphyrin (Protoporphyrin IX) within the structure of CAT@MnPFs can convert oxygen (O2) into singlet oxygen (1O2), showing the photodynamic effect. Importantly, the loaded catalase can decompose hydrogen peroxide (H2O2) into O2 with a huge elevation of O2 level (13.22 mg L-1) in 600 s, thus promoting 1O2 generation via PDT. As a result, CAT@MnPFs combined with 650 nm light can effectively ablate cancer cells due to the catalase-assisted oxygen-evolving PDT, showing a high therapeutic efficacy. Meanwhile, after the incubation with CAT@MnPFs, unobvious damage can be found in normal and red blood cells. Thus, the obtained CAT@MnPFs integrate the advantage of photosensitizers and catalase for oxygen-evolving PDT, which can provide some insight for treating hypoxic cells.
Collapse
Affiliation(s)
- Yang Qiao
- Department of Hematology and Oncology, Wenzhou Medical University affiliated Huangyan Hospital, The First People's Hospital of Taizhou, People's Republic of China
| | - Xiaowan Tang
- Department of Hematology and Oncology, Wenzhou Medical University affiliated Huangyan Hospital, The First People's Hospital of Taizhou, People's Republic of China
| | - Xu Qiuju
- The Third Affiliated Hospital of Harbin Medical University, 150 Haping Rd, Harbin, Heilongjiang Province, People's Republic of China
| | - Guangwen Zhang
- Department of Hematology and Oncology, Wenzhou Medical University affiliated Huangyan Hospital, The First People's Hospital of Taizhou, People's Republic of China
| |
Collapse
|
4
|
Zhao Q, Feng J, Liu F, Liang Q, Xie M, Dong J, Zou Y, Ye J, Liu G, Cao Y, Guo Z, Qiao H, Zheng L, Zhao K. Rhizoma Drynariae-derived nanovesicles reverse osteoporosis by potentiating osteogenic differentiation of human bone marrow mesenchymal stem cells via targeting ER α signaling. Acta Pharm Sin B 2024; 14:2210-2227. [PMID: 38799625 PMCID: PMC11119514 DOI: 10.1016/j.apsb.2024.02.005] [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: 11/26/2023] [Revised: 12/27/2023] [Accepted: 01/08/2024] [Indexed: 05/29/2024] Open
Abstract
Although various anti-osteoporosis drugs are available, the limitations of these therapies, including drug resistance and collateral responses, require the development of novel anti-osteoporosis agents. Rhizoma Drynariae displays a promising anti-osteoporosis effect, while the effective component and mechanism remain unclear. Here, we revealed the therapeutic potential of Rhizoma Drynariae-derived nanovesicles (RDNVs) for postmenopausal osteoporosis and demonstrated that RDNVs potentiated osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) by targeting estrogen receptor-alpha (ERα). RDNVs, a natural product isolated from fresh Rhizoma Drynariae root juice by differential ultracentrifugation, exhibited potent bone tissue-targeting activity and anti-osteoporosis efficacy in an ovariectomized mouse model. RDNVs, effectively internalized by hBMSCs, enhanced proliferation and ERα expression levels of hBMSC, and promoted osteogenic differentiation and bone formation. Mechanistically, via the ERα signaling pathway, RDNVs facilitated mRNA and protein expression of bone morphogenetic protein 2 and runt-related transcription factor 2 in hBMSCs, which are involved in regulating osteogenic differentiation. Further analysis revealed that naringin, existing in RDNVs, was the active component targeting ERα in the osteogenic effect. Taken together, our study identified that naringin in RDNVs displays exciting bone tissue-targeting activity to reverse osteoporosis by promoting hBMSCs proliferation and osteogenic differentiation through estrogen-like effects.
Collapse
Affiliation(s)
- Qing Zhao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Junjie Feng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fubin Liu
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qianxin Liang
- The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai 519000, China
| | - Manlin Xie
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiaming Dong
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yanfang Zou
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiali Ye
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Guilong Liu
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Department of Blood Transfusion, Guangdong Heyou International Hospital, Foshan 528306, China
| | - Yue Cao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhaodi Guo
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Hongzhi Qiao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lei Zheng
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Kewei Zhao
- Guangzhou Key Laboratory of Chinese Medicine Research on Prevention and Treatment of Osteoporosis, the Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510378, China
- Guangdong Engineering Research Center of Chinese Herbal-derived Vesicles, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
5
|
Ou R, Aodeng G, Ai J. Advancements in the Application of the Fenton Reaction in the Cancer Microenvironment. Pharmaceutics 2023; 15:2337. [PMID: 37765305 PMCID: PMC10536994 DOI: 10.3390/pharmaceutics15092337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/04/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Cancer is a complex and multifaceted disease that continues to be a global health challenge. It exerts a tremendous burden on individuals, families, healthcare systems, and society as a whole. To mitigate the impact of cancer, concerted efforts and collaboration on a global scale are essential. This includes strengthening preventive measures, promoting early detection, and advancing effective treatment strategies. In the field of cancer treatment, researchers and clinicians are constantly seeking new approaches and technologies to improve therapeutic outcomes and minimize adverse effects. One promising avenue of investigation is the utilization of the Fenton reaction, a chemical process that involves the generation of highly reactive hydroxyl radicals (·OH) through the interaction of hydrogen peroxide (H2O2) with ferrous ions (Fe2+). The generated ·OH radicals possess strong oxidative properties, which can lead to the selective destruction of cancer cells. In recent years, researchers have successfully introduced the Fenton reaction into the cancer microenvironment through the application of nanotechnology, such as polymer nanoparticles and light-responsive nanoparticles. This article reviews the progress of the application of the Fenton reaction, catalyzed by polymer nanoparticles and light-responsive nanoparticles, in the cancer microenvironment, as well as the potential applications and future development directions of the Fenton reaction in the field of tumor treatment.
Collapse
Affiliation(s)
| | | | - Jun Ai
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (R.O.); (G.A.)
| |
Collapse
|
6
|
Zhao L, Ren X, Ma H, Wang H, Li Y, Wei Q, Wu D, Ju H. Electrochemiluminescence Sensor with Controlled-Release Triggering Electrostatic Attraction Elimination Mechanism for Trenbolone Trace Detection. Anal Chem 2023; 95:13463-13469. [PMID: 37647570 DOI: 10.1021/acs.analchem.3c01359] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
A controlled-release strategy can meet the needs of sensitive environmental monitoring for pollutants through a self-on/off mode. In this work, an electrochemiluminescence (ECL) biosensor with controlled-release triggering electrostatic attraction elimination and biomolecular stimulated response strategies was constructed to detect environmental steroid hormones sensitively. The blocked pores on the aminated mesoporous silica nanocontainers were opened by specific binding between the trenbolone (TB) antigen and the antibody. The released l-cysteine counteracted the negative charge on the MnO2 NF surface through the redox reaction between -SH and MnO2, making the electrostatic interaction between the MnO2 NFs and the Ru(dcbpy)32+ disappear. Ru(dcbpy)32+ released an ECL signal on the electrode, thus completing the controlled-release triggering electrostatic attraction elimination strategy. In addition, with the TB antibody as the target and the competition strategy between the TB antigen and the standard substance, the constructed controlled-release ECL biosensor was used to detect the TB standard substance. Moreover, MnO2 NFs as the substrate of the ECL biosensor increased the active specific surface area of the electrode, effectively catalyzing the production of OH• and O2•-, thus endowing the ECL biosensor with coreactant-catalytic enhancement characteristic and further improving its ECL performance. This sensitive signal response brought about a low limit of detection of 2.53 fg/mL for the constructed ECL biosensor, which contributed a feasible idea for efficient trace analysis of pollutants in the environment.
Collapse
Affiliation(s)
- Lu Zhao
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Xiang Ren
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Hongmin Ma
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Huan Wang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Yuyang Li
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Qin Wei
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Dan Wu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Huangxian Ju
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| |
Collapse
|
7
|
Lin X, Cai L, Cao X, Zhao Y. Stimuli-responsive silk fibroin for on-demand drug delivery. SMART MEDICINE 2023; 2:e20220019. [PMID: 39188280 PMCID: PMC11235688 DOI: 10.1002/smmd.20220019] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/07/2022] [Indexed: 08/28/2024]
Abstract
Stimuli-responsive "smart" hydrogel biomaterials have attracted great attention in the biomedical field, especially in designing novel on-demand drug delivery systems. As a handful natural biomaterial approved by US Food and Drug Administration, silk fibroin (SF) has unique high temperature resistance as well as tunable structural composition. These properties make it one of the most ideal candidates for on-demand drug delivery. Meanwhile, recent advances in polymer modification and nanomaterials have fostered the development of various stimuli-responsive delivery systems. Here, we first review the recent advance in designing responsive SF-based delivery systems in different stimulus sources. These systems are able to release mediators in a desired manner in response to specific stimuli in active or passive manners. We then describe applications of these specially designed responsive delivery systems in wound healing, tumor therapy, as well as immunomodulation. We also discuss the future challenges and prospects of stimuli-responsive SF-based delivery systems.
Collapse
Affiliation(s)
- Xiang Lin
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical Engineering, Southeast UniversityNanjingChina
| | - Lijun Cai
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical Engineering, Southeast UniversityNanjingChina
| | - Xinyue Cao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical Engineering, Southeast UniversityNanjingChina
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalSchool of Biological Science and Medical Engineering, Southeast UniversityNanjingChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouChina
| |
Collapse
|
8
|
Fan H, Guo Z. Tumor microenvironment-responsive manganese-based nanomaterials for cancer treatment. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
9
|
Li B, Ye X, Fu Y, Feng L, Xu J, Niu X, Ye H, You Z. Hollow MnO 2-Based Nanoprobes for Enhanced Photothermal/Photodynamic /Chemodynamic Co-Therapy of Hepatocellular Carcinoma. Pharm Res 2023; 40:1271-1282. [PMID: 36991228 DOI: 10.1007/s11095-023-03501-4] [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: 09/27/2022] [Accepted: 03/14/2023] [Indexed: 03/31/2023]
Abstract
PURPOSE The effect of monotherapy in cancer is frequently influenced by the tumor's unique hypoxic microenvironment, insufficient drug concentration at the treatment site, and tumour cells' increased drug tolerance. In this work, we expect to design a novel therapeutic nanoprobe with the ability to solve these problems and improve the efficacy of antitumor therapy. METHODS We have prepared a hollow manganese dioxide nanoprobes loaded with photosensitive drug IR780 for the photothermal/photodynamic/chemodynamic co-therapy of liver cancer. RESULTS The nanoprobe demonstrates efficient thermal transformation ability under a single laser irradiation, and under the synergistic influence of photo heat, accelerates the Fenton/ Fenton-like reaction efficiency based on Mn2+ ions to produce more ·OH under the synergistic effect of photo heat. Moreover, the oxygen released under the degradation of manganese dioxide further promotes the ability of photosensitive drugs to produce singlet oxygen (ROS). The nanoprobe has been found to efficiently destroy tumour cells in vivo and in vitro experiments when used in combination with photothermal/photodynamic/ chemodynamic modes of treatment under laser irradiation. CONCLUSION In all, this research shows that a therapeutic strategy based on this nanoprobe could be a viable alternative for cancer treatment in the near future.
Collapse
Affiliation(s)
- Bei Li
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guoxue Lane, Wuhou District, Chengdu, 610041, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiwen Ye
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guoxue Lane, Wuhou District, Chengdu, 610041, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Lei Feng
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guoxue Lane, Wuhou District, Chengdu, 610041, China
| | - Jianrong Xu
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaoya Niu
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guoxue Lane, Wuhou District, Chengdu, 610041, China
| | - Hui Ye
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guoxue Lane, Wuhou District, Chengdu, 610041, China
| | - Zhen You
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, No.37 Guoxue Lane, Wuhou District, Chengdu, 610041, China.
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Chen C, Zhang W, Lu SY, Wang J, Tan Y, Zhao S, Ouyang Y, Xu L, Zhou B, Yin X, Ran H, Liu H. Repolarizing tumor-associated macrophages by layered double hydroxide-based deacidification agent for tumor chemodynamic therapy and immunotherapy. Colloids Surf B Biointerfaces 2023; 223:113157. [PMID: 36716658 DOI: 10.1016/j.colsurfb.2023.113157] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/23/2023]
Abstract
Tumor-associated macrophages (TAMs)-mediated immunotherapy has attracted extensive attention in tumor elimination. However, the acidic tumor microenvironment (TME) severely limits the phenotype of TAMs to pro-tumoral M2 state, suppressing immune response efficacy against tumors. Herein, novel poly(acrylic acid) (PAA)-coated, doxorubicin (DOX)-loaded layered double hydroxide (LDH) nanosheets (NSs) were developed as deacidification agent to repolarize TAMs from pro-tumoral M2 to anti-tumoral M1 phenotype for tumor elimination through combined chemodynamic therapy and immunotherapy. When located in tumor regions, LDH-PAA@DOX NSs display good deacidification capacity to neutralize acidic TME, achieving the repolarization of TAMs to M1 phenotype and further activating CD8+ T cells. During the deacidification process, these NSs are acid-responsive and degrade to release Fe3+ and DOX. The former can be reduced to Fe2+ by intracellular glutathione, meanwhile disrupting the antioxidant defense system of tumor cells. The latter can damage tumor cells directly and further stimulate the production of hydrogen peroxide, providing abundant substrate for the Fenton reaction. Toxic hydroxyl radical is excessively produced through Fe2+-mediated Fenton reaction to cause intratumoral oxidative stress. In vivo data revealed that significant tumor elimination can be achieved under LDH-PAA@DOX treatment. This work not only provides a promising paradigm for neutralizing acidic TME using deacidification agent but also highlights the effectiveness of combined chemodynamic therapy and immunotherapy in tumor treatment.
Collapse
Affiliation(s)
- Chunmei Chen
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Wei Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Shi-Yu Lu
- College of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Jingjing Wang
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Yixin Tan
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Sheng Zhao
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Yi Ouyang
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Luen Xu
- School of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Benqing Zhou
- Department of Biomedical Engineering, College of Engineering, Shantou University, Shantou 515063, China.
| | - Xuntao Yin
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China.
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China.
| | - Hui Liu
- School of Materials and Energy, Southwest University, Chongqing 400715, China.
| |
Collapse
|
11
|
Yu M, Cao R, Ma Z, Zhu M. Development of "smart" drug delivery systems for chemo/PDT synergistic treatment. J Mater Chem B 2023; 11:1416-1433. [PMID: 36734612 DOI: 10.1039/d2tb02248f] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Although chemotherapy and photodynamic therapy (PDT) have been developed for fighting cancer, the complex and heterogeneous nature of tumors makes it difficult for a single therapy to completely inhibit tumor growth. In order to reduce multidrug resistance of cancer cells to chemotherapeutic drugs and overcome low PDT efficiency in the hypoxic tumor microenvironment (TME), chemo/PDT synergistic treatment has received much attention in recent years. Depending on the characteristic signals of TME, various drug delivery systems can be constructed to target tumors and improve the therapeutic efficacy and the pharmacokinetic profile of anticancer drugs. This review highlights the synergistic strategies, treatment protocols, and design of chemo/PDT co-therapy in recent years to explore its scope and limitations. Taking advantage of stimuli-responsive materials and active cancer-targeting agents, cancer-targeting synergistic therapy is presented and discussed, providing ideas and suggestions for the construction of chemo/PDT co-therapy "smart" nanocarriers.
Collapse
Affiliation(s)
- Miaomiao Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China.
| | - Ran Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China.
| | - Zhiyuan Ma
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China.
| | - Meifang Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, 2999 North Renmin Road, Shanghai 201620, China.
| |
Collapse
|
12
|
Tumor Microenvironment-Responsive Magnetic Nanofluid for Enhanced Tumor MRI and Tumor multi-treatments. Pharmaceuticals (Basel) 2023. [DOI: 10.3390/ph16020166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
We prepared a tumor microenvironment-responsive magnetic nanofluid (MNF) for improving tumor targeting, imaging and treatment simultaneously. For this purpose, we synthesized sulfonamide-based amphiphilic copolymers with a suitable pKa at 7.0; then, we utilized them to prepare the tumor microenvironment-responsive MNF by self-assembly of the sulfonamide-based amphiphilic copolymers and hydrophobic monodispersed Fe3O4 nanoparticles at approximately 8 nm. After a series of characterizations, the MNF showed excellent application potential due to the fact of its high stability under physiological conditions and its hypersensitivity toward tumor stroma by forming aggregations within neutral or weak acidic environments. Due to the fact of its tumor microenvironment-responsiveness, the MNF showed great potential for accumulation in tumors, which could enhance MNF-mediated magnetic resonance imaging (MRI), magnetic hyperthermia (MH) and Fenton reaction (FR) in tumor. Moreover, in vitro cell experiment did not only show high biocompatibility of tumor microenvironment-responsive MNF in physiological environment, but also exhibit high efficacy on inhibiting cell proliferation by MH-dependent chemodynamic therapy (CDT), because CDT was triggered and promoted efficiently by MH with increasing strength of alternating magnetic field. Although the current research is limited to in vitro study, these positive results still suggest the great potential of the MNF on effective targeting, diagnosis, and therapy of tumor.
Collapse
|
13
|
Qiu X, Li Z, Wu Y, Binder WH, Chen S, Zhu J. Core-Coordinated Elliptic Polymer Nanoparticles Loading Copper(II) and Chlorambucil for Cooperative Chemodynamic/Chemotherapy. Biomacromolecules 2022; 23:4519-4531. [PMID: 36250649 DOI: 10.1021/acs.biomac.2c00656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Chemodynamic therapy (CDT) reflects an innovative cancer treatment modality; however, to enhance its relatively low therapeutic efficiency, rational combination with extra therapeutic modes is highly appreciated. Here, core-coordinated amphiphilic, elliptic polymer nanoparticles (Cu/CBL-POEGEA NPs) are constructed via the self-assembly of a glutathione (GSH)-responsive polymer-drug conjugate, bearing side-chain acylthiourea (ATU) motifs which behave as ligands capable of coordinating Cu(II), such a design is featured by combined chemo (CT)/CDT with dual GSH depletion collectively triggered by the Cu(II) reduction reaction and disulfide bond breakage. To do so, an amphiphilic random copolymer poly[oligo(ethylene glycol)ethyl acrylate-co-thiourea] [P(OEGEA-co-ATU)] is synthesized, followed by conjugation of chlorambucil (CBL) to ATU motifs linked via a disulfide bond, thus yielding the targeted P[OEGEA-co-(ATU-g-CBL)]. In such a system, hydrophilic POEGEA serves as the biocompatible section and ATU motifs coordinate Cu(II), resulting in core-coordinated elliptic Cu/CBL-POEGEA NPs. Benefitting from the GSH-induced reduction reaction, Cu(II) is converted into Cu(I) and subsequently react with endogenous H2O2 to create •OH, realizing GSH-depletion-promoted CDT. Additionally, the disulfide bond endows GSH-responsive CBL release and provokes further GSH decline, finally realizing combined CDT/CT toward enhancing antitumor outcomes, and in vitro as well as in vivo studies indeed reveal remarkable efficacy. Such a system can provide valuable advantages to create novel nanomedicines toward cascade antitumor therapy.
Collapse
Affiliation(s)
- Xiaoyang Qiu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zeke Li
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yanggui Wu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wolfgang H Binder
- Chair of Macromolecular Chemistry, Faculty of Natural Science II (Chemistry, Physics and Mathematics), Martin Luther University Halle-Wittenberg, von-Danckelmann-Platz 4, Halle (Saale) D-06120, Germany
| | - Senbin Chen
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jintao Zhu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
14
|
Han W, Wei Z, Feng L, Yao M, Zhang H, Zhang S. Single-Site Fe-N-C Atom Based Carbon Nanotubes for Mutually Promoted and Synergistic Oncotherapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:48356-48367. [PMID: 36281918 DOI: 10.1021/acsami.2c11809] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
A carbon nanotube (CNT) supported single-site Fe-N-C catalyst (CNTs/Fe-N-C) exhibited attractive properties in peroxidase (POD)-like activity and photothermal effect. Herein, we designed a therapeutic platform by wrapping doxorubicin (DOX) in mesoporous CNTs/Fe-N-C with the cell membrane (CM) of breast cancer. The ultimate nanoagent (CNTs/Fe-N-C/DOX/CM) exhibited high antitumor activity on account of its efficient catalytic ability, increased drug release rates, and significant photothermal effect. Due to the POD-like activity, CNTs/Fe-N-C could effectively catalyze hydrogen peroxide (H2O2) into cytotoxic hydroxyl radicals (•OH) for chemodynamic therapy (CDT) of the tumor. Besides, the released DOX not only merely induced the diagnosis of the tumor cells for chemotherapy (CT) but also generated H2O2 to promote CDT. Moreover, the photothermal effect of the nanoagent could use for photothermal therapy (PTT). The increasing temperature was conducive to the release of DOX from micropore into the cell, which indirectly enhanced CT and CDT effects. As an intelligent and multifunctional drug delivery platform, the present CNTs/Fe-N-C/DOX/CM nanoagent could be engineered with synergistic treatments and favorable biosafety, which provides a promising paradigm in site-specific antitumor treatment and biomedicine.
Collapse
Affiliation(s)
- Wenxiu Han
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Makers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, People's Republic of China
| | - Zizhen Wei
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Makers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, People's Republic of China
| | - Lu Feng
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Makers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, People's Republic of China
| | - Mei Yao
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Makers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, People's Republic of China
| | - Huairong Zhang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Makers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, People's Republic of China
| | - Shusheng Zhang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Makers, Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, People's Republic of China
| |
Collapse
|
15
|
Qiu M, Chen J, Huang X, Li B, Zhang S, Liu P, Wang Q, Qian ZR, Pan Y, Chen Y, Zhao J. Engineering Chemotherapeutic-Augmented Calcium Phosphate Nanoparticles for Treatment of Intraperitoneal Disseminated Ovarian Cancer. ACS APPLIED MATERIALS & INTERFACES 2022; 14:21954-21965. [PMID: 35508299 DOI: 10.1021/acsami.2c02552] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Ovarian cancer is a common gynecologic malignancy with a high fatality rate. Intraperitoneal chemotherapy has been proved as an efficient clinical treatment for disseminated ovarian cancer. However, there are limitations for conventional small molecule drugs to achieve an ideal therapeutic effect. Herein, a synergistic treatment for intraperitoneally disseminated ovarian cancer was achieved by Arg-Gly-Asp (RGD)-modified amorphous calcium phosphate loading with doxorubicin (designated as RGD-CaPO/DOX). The engineered calcium-involved nanomedicine augmented the therapeutic effect of DOX by aggravating endoplasmic reticulum stress, calcium overload, and mitochondrial dysfunction, ultimately triggering mitochondrial apoptosis in the SKOV3 (human ovarian cancer) cell line. In an intraperitoneally disseminated tumor model, RGD modification and the weak negative surface potential of the NPs were beneficial for intraperitoneal retention and tumor targeting. Moreover, intraperitoneal injection of RGD-CaPO/DOX NPs resulted in a favorable antitumor effect. The mean survival time of SKOV3-bearing mice was significantly extended from 29 to 59 days with negligible toxicity. Therefore, this study has been designed to provide an effective chemotherapeutic-augmented treatment for intraperitoneally disseminated ovarian cancer.
Collapse
Affiliation(s)
- Miaojuan Qiu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Junzong Chen
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Xiuyu Huang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Binbin Li
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Shiqiang Zhang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Peng Liu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Qiang Wang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Zhi Rong Qian
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Yihang Pan
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Jing Zhao
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, P. R. China
| |
Collapse
|
16
|
Zhang X, He Q, Sun J, Gong H, Cao Y, Duan L, Yi S, Ying B, Xiao B. Near-Infrared-Enpowered Nanomotor-Mediated Targeted Chemotherapy and Mitochondrial Phototherapy to Boost Systematic Antitumor Immunity. Adv Healthc Mater 2022; 11:e2200255. [PMID: 35536883 DOI: 10.1002/adhm.202200255] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/02/2022] [Indexed: 02/05/2023]
Abstract
Phototherapy is an important strategy to inhibit tumor growth and activate antitumor immunity. However, the effect of photothermal/photodynamic therapy (PTT/PDT) is restricted by limited tumor penetration depth and unsatisfactory potentiation of antitumor immunity. Here, a near-infrared (NIR)-driven nanomotor is constructed with a mesoporous silicon nanoparticle (MSN) as the core, end-capped with Antheraea pernyi silk fibroin (ApSF) comprising arginine-glycine-aspartate (RGD) tripeptides. Upon NIR irradiation, the resulting ApSF-coated MSNs (DIMs) loading with photosensitizers (ICG derivatives, IDs) and chemotherapeutic drugs (doxorubicin, Dox) can efficiently penetrate into the internal tumor tissues and achieve effective phototherapy. Combined with chemotherapy, a triple-modal treatment (PTT, PDT, and chemotherapy) approach is developed to induce the immunogenic cell death of tumor cells and to accelerate the release of damage-associated molecular patterns. In vivo results suggest that DIMs can promote the maturation of dendritic cells and surge the number of infiltrated immune cells. Meanwhile, DIMs can polarize macrophages from M2 to M1 phenotypes and reduce the percentages of immunosuppressive Tregs, which reverse the immunosuppressive tumor microenvironment and activate systemic antitumor immunity. By achieving synergistic effects on the tumor inhibition and the antitumor immunity activation, DIMs show great promise as new nanoplatforms to treat metastatic breast cancer.
Collapse
Affiliation(s)
- Xueqing Zhang
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass Sciences Southwest University Chongqing 400715 China
| | - Qian He
- West China Hospital Sichuan University Chengdu 610041 China
| | - Jianfeng Sun
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences University of Oxford Headington Oxford OX3 7LD UK
| | - Hanlin Gong
- West China Hospital Sichuan University Chengdu 610041 China
| | - Yingui Cao
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass Sciences Southwest University Chongqing 400715 China
| | - Lian Duan
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass Sciences Southwest University Chongqing 400715 China
| | - Shixiong Yi
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass Sciences Southwest University Chongqing 400715 China
| | - Binwu Ying
- West China Hospital Sichuan University Chengdu 610041 China
| | - Bo Xiao
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass Sciences Southwest University Chongqing 400715 China
| |
Collapse
|
17
|
Wang Y, Huo J, Li S, Huang R, Fan D, Cheng H, Wan B, Du Y, He H, Zhang G. Self-Rectifiable and Hypoxia-Assisted Chemo-Photodynamic Nanoinhibitor for Synergistic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:10092-10101. [PMID: 35170301 DOI: 10.1021/acsami.1c23121] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Photodynamic therapy (PDT) can eradicate cancer cells under light irradiation, mainly because of reactive singlet oxygen (1O2) being transformed from intratumoral oxygen. Nonetheless, the consumption of oxygen during PDT results in serious hypoxic conditions and an elevated hypoxia-inducing factor-1α (HIF-1α) level that hamper further photodynamic efficacy and induce tumor metastasis. To address this problem, we developed hypoxia-assisted NP-co-encapsulating Ce6 (photosensitizer) and YC-1 (HIF-1α inhibitor) as a self-rectifiable nanoinhibitor for synergistic antitumor treatment. PDT-aggravated intracellular hypoxic stress facilitated NP dissociation to release the drug (YC-1), which achieved tumor killing and HIF-1α inhibition to further enhance the therapeutic effect of PDT and prevent tumor metastasis. Besides, in vivo studies revealed that the HC/PI@YC-1 NPs afforded synergistic anticancer efficacy with minimal toxicity. Therefore, this study provides a prospective approach against PDT drawbacks and combination cancer therapy.
Collapse
Affiliation(s)
- Yanan Wang
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Jian Huo
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Shuang Li
- Department of Pathology, Zhengzhou University People's Hospital (Henan Provincial People's Hospital), Zhengzhou 450003, China
| | - Ran Huang
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Daopeng Fan
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Hanghang Cheng
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Bo Wan
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Yongkun Du
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Hua He
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| | - Gaiping Zhang
- College of Veterinary Medicine, International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou 450046, China
| |
Collapse
|
18
|
Han S, Yao A, Ding Y, Leng Q, Teng F, Zhao L, Sun R, Bu H. A dual-template imprinted polymer based on amino-functionalized zirconium-based metal-organic framework for delivery of doxorubicin and phycocyanin with synergistic anticancer effect. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111161] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
19
|
Chen Q, Li Q, Liang Y, Zu M, Chen N, Canup BS, Luo L, Wang C, Zeng L, Xiao B. Natural exosome-like nanovesicles from edible tea flowers suppress metastatic breast cancer via ROS generation and microbiota modulation. Acta Pharm Sin B 2022; 12:907-923. [PMID: 35256954 PMCID: PMC8897038 DOI: 10.1016/j.apsb.2021.08.016] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/22/2021] [Accepted: 07/05/2021] [Indexed: 12/17/2022] Open
Abstract
Although several artificial nanotherapeutics have been approved for practical treatment of metastatic breast cancer, their inefficient therapeutic outcomes, serious adverse effects, and high cost of mass production remain crucial challenges. Herein, we developed an alternative strategy to specifically trigger apoptosis of breast tumors and inhibit their lung metastasis by using natural nanovehicles from tea flowers (TFENs). These nanovehicles had desirable particle sizes (131 nm), exosome-like morphology, and negative zeta potentials. Furthermore, TFENs were found to contain large amounts of polyphenols, flavonoids, functional proteins, and lipids. Cell experiments revealed that TFENs showed strong cytotoxicities against cancer cells due to the stimulation of reactive oxygen species (ROS) amplification. The increased intracellular ROS amounts could not only trigger mitochondrial damage, but also arrest cell cycle, resulting in the in vitro anti-proliferation, anti-migration, and anti-invasion activities against breast cancer cells. Further mice investigations demonstrated that TFENs after intravenous (i.v.) injection or oral administration could accumulate in breast tumors and lung metastatic sites, inhibit the growth and metastasis of breast cancer, and modulate gut microbiota. This study brings new insights to the green production of natural exosome-like nanoplatform for the inhibition of breast cancer and its lung metastasis via i.v. and oral routes.
Collapse
Key Words
- AF633, Alexa Fluor 633-labeled phalloidin
- ALP, alkaline phosphatase
- ALT, alanine aminotransferase
- AST, aspartate aminotransferase
- BUN, urea nitrogen
- Breast cancer
- CDK, CYCLIN-dependent kinase
- CRE, creatinine
- DAF-FM DA, 4-amino-5-methylamino-2′,7′-difluorofluorescein diacetate
- DAPI, 4′,6-diamidino-2-phenylindole
- DCFH-DA, dichloro-dihydro-fluorescein diacetate
- DGDG, digalactosyl diacylglycerols
- DHE, dihydroethidium
- DLS, dynamic light scattering
- DiO, 3,3′-dioctadecyloxacarbocyanine perchlorate
- DiR, 1,1′-dioctadecyl-3,3,3′′,3′-tetramethylindotricarbocyanine iodide
- EC, epicatechin
- ECG, epicatechin gallate
- EGCG, epigallocatechin gallate
- Exosome-like nanoparticle
- FBS, fetal bovine serum
- GIT, gastrointestinal tract
- H&E, Hematoxylin & Eosin
- HPLC, high-performance liquid chromatography
- Intravenous injection
- LC‒MS, liquid chromatography‒mass spectrometry
- MFI, mean fluorescence intensity
- MGDG, monogalactosyl diacylglycerols
- MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- Metastasis
- Microbiota modulation
- NO, nitrogen monoxide
- NPs, nanoparticles
- OUT, operational taxonomic unit
- Oral administration
- PA, phosphatidic acids
- PBS, phosphate-buffered saline
- PC, phosphatidylcholines
- PDI, polydispersity index
- PE, phosphatidylethanolamines
- PG, phosphatidylglycerol
- PI, phosphatidylinositol
- PLT, platelets
- PMe, phosphatidylmethanol
- PS, phosphatidylserine
- RBC, red blood cell
- RNS, reactive nitrogen species
- ROS generation
- ROS, reactive oxygen species
- SA, superoxide anion
- SQDG, sulphoquinovosyl diylyceride
- TEM, transmission electron microscopy
- TFENs, exosome-like NPs from tea flowers
- TG, triglyceride
- TUNEL, TdT-mediated dUTP Nick-end labeling
- Tea flower
- WBC, white blood cell
Collapse
|
20
|
Jia C, Guo Y, Wu FG. Chemodynamic Therapy via Fenton and Fenton-Like Nanomaterials: Strategies and Recent Advances. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2103868. [PMID: 34729913 DOI: 10.1002/smll.202103868] [Citation(s) in RCA: 233] [Impact Index Per Article: 116.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/23/2021] [Indexed: 06/13/2023]
Abstract
Chemodynamic therapy (CDT), a novel cancer therapeutic strategy defined as the treatment using Fenton or Fenton-like reaction to produce •OH in the tumor region, was first proposed by Bu, Shi, and co-workers in 2016. Recently, with the rapid development of Fenton and Fenton-like nanomaterials, CDT has attracted tremendous attention because of its unique advantages: 1) It is tumor-selective with low side effects; 2) the CDT process does not depend on external field stimulation; 3) it can modulate the hypoxic and immunosuppressive tumor microenvironment; 4) the treatment cost of CDT is low. In addition to the Fe-involved CDT strategies, the Fenton-like reaction-mediated CDT strategies have also been proposed, which are based on many other metal elements including copper, manganese, cobalt, titanium, vanadium, palladium, silver, molybdenum, ruthenium, tungsten, cerium, and zinc. Moreover, CDT has been combined with other therapies like chemotherapy, radiotherapy, phototherapy, sonodynamic therapy, and immunotherapy for achieving enhanced anticancer effects. Besides, there have also been studies that extend the application of CDT to the antibacterial field. This review introduces the latest advancements in the nanomaterials-involved CDT from 2018 to the present and proposes the current limitations as well as future research directions in the related field.
Collapse
Affiliation(s)
- Chenyang Jia
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Yuxin Guo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| |
Collapse
|
21
|
Ma Y, Duan L, Sun J, Gou S, Chen F, Liang Y, Dai F, Xiao B. Oral nanotherapeutics based on Antheraea pernyi silk fibroin for synergistic treatment of ulcerative colitis. Biomaterials 2022; 282:121410. [DOI: 10.1016/j.biomaterials.2022.121410] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/29/2022] [Accepted: 02/13/2022] [Indexed: 01/08/2023]
|
22
|
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
|
23
|
Jin ZY, Fatima H, Zhang Y, Shao Z, Chen XJ. Recent Advances in Bio‐Compatible Oxygen Singlet Generation and Its Tumor Treatment. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Zheng Yang Jin
- The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| | - Hira Fatima
- Western Australia School of Mines: Minerals Energy and Chemical Engineering (WASM‐MECE) Curtin University Perth Western Australia 6102 Australia
| | - Yue Zhang
- The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| | - Zongping Shao
- Western Australia School of Mines: Minerals Energy and Chemical Engineering (WASM‐MECE) Curtin University Perth Western Australia 6102 Australia
- State Key Laboratory of Materials‐Oriented Chemical Engineering College of Chemical Engineering Nanjing Tech University Nanjing Jiangsu 211816 P. R. China
| | - Xiang Jian Chen
- The First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325015 P. R. China
| |
Collapse
|
24
|
Zhou QM, Lu YF, Zhou JP, Yang XY, Wang XJ, Yu JN, Du YZ, Yu RS. Self-amplification of oxidative stress with tumour microenvironment-activatable iron-doped nanoplatform for targeting hepatocellular carcinoma synergistic cascade therapy and diagnosis. J Nanobiotechnology 2021; 19:361. [PMID: 34749740 PMCID: PMC8576982 DOI: 10.1186/s12951-021-01102-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 10/21/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma is insensitive to many chemotherapeutic agents. Ferroptosis is a form of programmed cell death with a Fenton reaction mechanism. It converts endogenous hydrogen peroxide into highly toxic hydroxyl radicals, which inhibit hepatocellular carcinoma progression. METHODS The morphology, elemental composition, and tumour microenvironment responses of various organic/inorganic nanoplatforms were characterised by different analytical methods. Their in vivo and in vitro tumour-targeting efficacy and imaging capability were analysed by magnetic resonance imaging. Confocal microscopy, flow cytometry, and western blotting were used to investigate the therapeutic efficacy and mechanisms of complementary ferroptosis/apoptosis mediated by the nanoplatforms. RESULTS The nanoplatform consisted of a silica shell doped with iron and disulphide bonds and an etched core loaded with doxorubicin that generates hydrogen peroxide in situ and enhances ferroptosis. It relied upon transferrin for targeted drug delivery and could be activated by the tumour microenvironment. Glutathione-responsive biodegradability could operate synergistically with the therapeutic interaction between doxorubicin and iron and induce tumour cell death through complementary ferroptosis and apoptosis. The nanoplatform also has a superparamagnetic framework that could serve to guide and monitor treatment under T2-weighted magnetic resonance imaging. CONCLUSION This rationally designed nanoplatform is expected to integrate cancer diagnosis, treatment, and monitoring and provide a novel clinical antitumour therapeutic strategy.
Collapse
Affiliation(s)
- Qiao-Mei Zhou
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang Province, People's Republic of China
| | - Yuan-Fei Lu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang Province, People's Republic of China
| | - Jia-Ping Zhou
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang Province, People's Republic of China
| | - Xiao-Yan Yang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang Province, People's Republic of China
| | - Xiao-Jie Wang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang Province, People's Republic of China
| | - Jie-Ni Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang Province, People's Republic of China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China.
| | - Ri-Sheng Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang Province, People's Republic of China.
| |
Collapse
|
25
|
Blum NT, Fu LH, Lin J, Huang P. When Chemodynamic Therapy Meets Photodynamic Therapy: A Synergistic Combination of Cancer Treatments. IEEE NANOTECHNOLOGY MAGAZINE 2021. [DOI: 10.1109/mnano.2021.3081755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
26
|
Smith CB, Days LC, Alajroush DR, Faye K, Khodour Y, Beebe SJ, Holder AA. Photodynamic Therapy of Inorganic Complexes for the Treatment of Cancer †. Photochem Photobiol 2021; 98:17-41. [PMID: 34121188 DOI: 10.1111/php.13467] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/10/2021] [Indexed: 01/05/2023]
Abstract
Photodynamic therapy (PDT) is a medicinal tool that uses a photosensitizer and a light source to treat several conditions, including cancer. PDT uses reactive oxygen species such as cytotoxic singlet oxygen (1 O2 ) to induce cell death in cancer cells. Chemotherapy has historically utilized the cytotoxic effects of many metals, especially transition metal complexes. However, chemotherapy is a systemic treatment so all cells in a patient's body are exposed to the same cytotoxic effects. Transition metal complexes have also shown high cytotoxicity as PDT agents. PDT is a potential localized method for treating several cancer types by using inorganic complexes as photosensitizing agents. This review covers several in vitro and in vivo studies, as well as clinical trials that reported on the anticancer properties of inorganic pharmaceuticals used in PDT against different types of cancer.
Collapse
Affiliation(s)
- Chloe B Smith
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA
| | - Lindsay C Days
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA
| | - Duaa R Alajroush
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA
| | - Khadija Faye
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA
| | - Yara Khodour
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA
| | - Stephen J Beebe
- Frank Reidy Research Centre for Bioelectrics, Old Dominion University, Norfolk, VA
| | - Alvin A Holder
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA
| |
Collapse
|