1
|
Bu JW, Wang ZG, Liu HY, Liu SL. Metal nanozymes modulation of reactive oxygen species as promising strategies for cancer therapy. Int J Pharm 2024; 662:124453. [PMID: 39013531 DOI: 10.1016/j.ijpharm.2024.124453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/11/2024] [Accepted: 07/07/2024] [Indexed: 07/18/2024]
Abstract
Nanozymes, nanostructured materials emulating natural enzyme activities, exhibit potential in catalyzing reactive oxygen species (ROS) production for cancer treatment. By facilitating oxidative reactions, elevating ROS levels, and influencing the tumor microenvironment (TME), nanozymes foster the eradication of cancer cells. Noteworthy are their superior stability, ease of preservation, and cost-effectiveness compared to natural enzymes, rendering them invaluable for medical applications. This comprehensive review intricately explores the interplay between ROS and tumor therapy, with a focused examination of metal-based nanozyme strategies mitigating tumor hypoxia. It provides nuanced insights into diverse catalytic processes, mechanisms, and surface modifications of various metal nanozymes, shedding light on their role in intra-tumoral ROS generation and applications in antioxidant therapy. The review concludes by delineating specific potential prospects and challenges associated with the burgeoning use of metal nanozymes in future tumor therapies.
Collapse
Affiliation(s)
- Jin-Wei Bu
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, P. R. China
| | - Zhi-Gang Wang
- College of Chemistry and School of Medicine, Nankai University, Tianjin 300071, P. R. China
| | - Hao-Yang Liu
- College of Chemistry and School of Medicine, Nankai University, Tianjin 300071, P. R. China.
| | - Shu-Lin Liu
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, P. R. China; College of Chemistry and School of Medicine, Nankai University, Tianjin 300071, P. R. China.
| |
Collapse
|
2
|
Zhou Y, Kou J, Zhang Y, Ma R, Wang Y, Zhang J, Zhang C, Zhan W, Li K, Li X. Magnetic-guided nanocarriers for ionizing/non-ionizing radiation synergistic treatment against triple-negative breast cancer. Biomed Eng Online 2024; 23:67. [PMID: 39003472 PMCID: PMC11245775 DOI: 10.1186/s12938-024-01263-7] [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: 05/01/2024] [Accepted: 07/03/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a subtype of breast cancer with the worst prognosis. Radiotherapy (RT) is one of the core modalities for the disease; however, the ionizing radiation of RT has severe side effects. The consistent development direction of RT is to achieve better therapeutic effect with lower radiation dose. Studies have demonstrated that synergistic effects can be achieved by combining RT with non-ionizing radiation therapies such as light and magnetic therapy, thereby achieving the goal of dose reduction and efficacy enhancement. METHODS In this study, we applied FeCo NPs with magneto thermal function and phototherapeutic agent IR-780 to construct an ionizing and non-ionizing radiation synergistic nanoparticle (INS NPs). INS NPs are first subjected to morphology, size, colloidal stability, loading capacity, and photothermal conversion tests. Subsequently, the cell inhibitory and cellular internalization were evaluated using cell lines in vitro. Following comprehensive assessment of the NPs' in vivo biocompatibility, tumor-bearing mouse model was established to evaluate their distribution, targeted delivery, and anti-tumor effects in vivo. RESULTS INS NPs have a saturation magnetization exceeding 72 emu/g, a hydrodynamic particle size of approximately 40 nm, a negatively charged surface, and good colloidal stability and encapsulation properties. INS NPs maintain the spectral characteristics of IR-780 at 808 nm. Under laser irradiation, the maximum temperature was 92 °C, INS NPs also achieved the effective heat temperature in vivo. Both in vivo and in vitro tests have proven that INS NPs have good biocompatibility. INS NPs remained effective for more than a week after one injection in vivo, and can also be guided and accumulated in tumors through permanent magnets. Later, the results exhibited that under low-dose RT and laser irradiation, the combined intervention group showed significant synergetic effects, and the ROS production rate was much higher than that of the RT and phototherapy-treated groups. In the mice model, 60% of the tumors were completely eradicated. CONCLUSIONS INS NPs effectively overcome many shortcomings of RT for TNBC and provide experimental basis for the development of novel clinical treatment methods for TNBC.
Collapse
Affiliation(s)
- Yun Zhou
- College of Clinical Medicine, Xi'an Medical University, Xi'an, 710021, China
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, China
| | - Junhao Kou
- College of Pharmacy, Xi'an Medical University, Xi'an, 710021, China
| | - Yuhuang Zhang
- College of Clinical Medicine, Xi'an Medical University, Xi'an, 710021, China
| | - Rongze Ma
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yao Wang
- Department of Radiation Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Junfeng Zhang
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, China
| | - Chunhong Zhang
- Xi'an Key Laboratory of Advanced Control and Intelligent Process, School of Automation, Xi'an University of Posts & Telecommunications, Xi'an, 710121, China
| | - Wenhua Zhan
- Department of Radiotherapy, General Hospital of Ningxia Medical University, Yinchuan, 750004, China.
| | - Ke Li
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, China.
| | - Xueping Li
- College of Clinical Medicine, Xi'an Medical University, Xi'an, 710021, China.
- Xi'an Key Laboratory for Prevention and Treatment of Common Aging Diseases, Translational and Research Centre for Prevention and Therapy of Chronic Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, China.
| |
Collapse
|
3
|
Cui W, Zhu S, Pan X, Zhang W, Wang T. Gold(III) Porphyrin-Metal-Polyphenolic Nanocomplexes: Breaking Intracellular Redox Environment for Enhancing Mild-Temperature Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30810-30818. [PMID: 38850233 DOI: 10.1021/acsami.4c04196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2024]
Abstract
Photothermal therapy (PTT) is a promising clinical antitumor strategy. However, local hyperthermia inevitably induces heat damage to adjacent normal tissues, while alternative mild-temperature therapy (MPTT, T < 45 °C) is also inefficient due to the overexpressed hyperthermia-induced heat shock proteins (HSPs) by cancer cells. Therefore, developing PTT strategies with minimizing damage to healthy tissues with improved cellular temperature sensitivity is extremely valuable for clinical application. Herein, we proposed the strategy of disrupting the intracellular redox environment via destroying the ROS-defending systems to promote MPTT. The gold(III) porphyrin-Fe3+-tannic acid nanocomplexes (AuTPP@TA-Fe NPs) were achieved via interfacial cohesion and supramolecular assembly of bioadhesive species, which could trigger the Fenton reaction to produce ·OH radicals and downregulation of reductive TrxR enzyme and mitochondrial chaperone protein Hsp60. The aggravation of oxides and the inactivation of Hsp60 provide favorable pathways for impeding the heat shock-induced self-repair mechanism of cancer cells, which strengthens AuTPP@TA-Fe NPs mediated MPTT.
Collapse
Affiliation(s)
- Weiwei Cui
- Life and Health Intelligent Research Institute, Tianjin Key Laboratory of Life and Health Detection, Tianjin University of Technology, Tianjin 300384, P. R. China
| | - Shan Zhu
- Life and Health Intelligent Research Institute, Tianjin Key Laboratory of Life and Health Detection, Tianjin University of Technology, Tianjin 300384, P. R. China
| | - Xiangmei Pan
- Life and Health Intelligent Research Institute, Tianjin Key Laboratory of Life and Health Detection, Tianjin University of Technology, Tianjin 300384, P. R. China
| | - Wei Zhang
- Life and Health Intelligent Research Institute, Tianjin Key Laboratory of Life and Health Detection, Tianjin University of Technology, Tianjin 300384, P. R. China
| | - Tie Wang
- Life and Health Intelligent Research Institute, Tianjin Key Laboratory of Life and Health Detection, Tianjin University of Technology, Tianjin 300384, P. R. China
| |
Collapse
|
4
|
Hu X, Li R, Liu J, Fang K, Dong C, Shi S. Engineering Dual-Responsive Prodrug-MOFs as Immunogenic Cell Death Initiator for Enhancing Cancer Immunotherapy. Adv Healthc Mater 2024; 13:e2302333. [PMID: 38253350 DOI: 10.1002/adhm.202302333] [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: 07/22/2023] [Revised: 12/10/2023] [Indexed: 01/24/2024]
Abstract
In recent years, the anticancer effects of disulfiram, a clinical drug for anti-alcoholism, are confirmed. However, several defects limit the clinical translation of disulfiram obviously, such as Cu(II)-dependent anticancer activity, instability, and non-selectivity for cancer cells. Herein, a phosphate and hydrogen peroxide dual-responsive nanoplatform (PCu-HA-DQ) is reported, which is constructed by encapsulating disulfiram prodrug (DQ) and modifying hyaluronic acid (HA) on copper doping metal-organic frameworks (PCu MOFs). PCu-HA-DQ is expected to accumulate in tumor by targeting CD-44 receptors and enable guidance with magnetic resonance imaging. Inside the tumor, Cu(DTC)2 will be generated in situ based on a dual-responsive reaction. In detail, the high concentration of phosphate can induce the release of DQ, after that, the intracellular hydrogen peroxide will further mediate the generation of Cu(DTC)2 . In vitro and in vivo results indicate PCu-HA-DQ can induce the apoptosis as well as immunogenic cell death (ICD) of tumor cells distinctly, leading to enhanced immune checkpoint inhibitor (ICI) efficacy by combining the anti-programmed death-1 antibody. This work provides a portable strategy to construct a dual-responsive nanoplatform integrating tumor-targeted ability and multi-therapy, and the designed nanoplatform is also an ICD inducer, which presents a prospect for boosting systemic antitumor immunity and ICI efficacy.
Collapse
Affiliation(s)
- Xiaochun Hu
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Department of Comprehensive Cancer Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
- School of Medicine, Shanghai University, Shanghai, 200444, P. R. China
| | - Ruihao Li
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Department of Comprehensive Cancer Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Jie Liu
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Department of Comprehensive Cancer Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Kang Fang
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Department of Comprehensive Cancer Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Chunyan Dong
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Department of Comprehensive Cancer Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| | - Shuo Shi
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Department of Comprehensive Cancer Therapy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, P. R. China
| |
Collapse
|
5
|
Xu Y, Yang L, Wang C, Sun W, Zheng Y, Ou B, Wu L, Shi L, Lin X, Chen W. Ferroptosis boosted oral cancer photodynamic therapy by carrier-free Sorafenib-Ce6 self-assembly nanoparticles. J Control Release 2024; 366:798-811. [PMID: 38184236 DOI: 10.1016/j.jconrel.2023.12.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/19/2023] [Accepted: 12/31/2023] [Indexed: 01/08/2024]
Abstract
Oral cancer is a disease with high morbidity and mortality worldwide and greatly impacts the quality of life, especially in patients with advanced stages. Photodynamic therapy (PDT) is one of the most effective clinical treatments for oral cancers. However, most clinically applied photosensitizers have several deficiencies, including oxygen dependence, poor aqueous solubility, and a lack of tumor-targeting ability. Herein, the carrier-free multifunctional Sorafenib (Sor), chlorin e6 (Ce6), and Fe3+ self-assembly co-delivery nanoparticles (Sor-Ce6 NPs) were constructed via combining a ferroptosis inducer Sor and a photosensitizer Ce6 for synergetic therapy. The as-synthesized Sor-Ce6 NPs presented excellent colloidal stability and water dispersity with good in vivo tumor-targeting ability. More significantly, the low dose of Sor-Ce6 NPs had little dark toxicity but produced significantly enhanced ROS and supplied O2 sustainably to increase phototoxicity through ferroptosis pathway. Notably, the Sor-Ce6 NPs showed significantly higher in vitro and in vivo anti-tumor efficacy than the Sor/Ce6 mixture due to the improvement of cellular uptake and the incorporation of foreign Fe ions in the system, which also confer the T1 magnetic resonance-guided imaging ability to the formed Sor-Ce6 NPs. Our study demonstrates a promising self-assembled strategy for overcoming hypoxia-related PDT resistance for oral cancer treatment.
Collapse
Affiliation(s)
- Yingying Xu
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Liu Yang
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Chengyan Wang
- Laboratory Animal Center, Fujian Medical University, Fuzhou 350122, China
| | - Weiming Sun
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Yijing Zheng
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Beiwei Ou
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Lixian Wu
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Leilei Shi
- Precision Research Center for Refractory Diseases in Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Xi Lin
- Public Technology Service Center, Fujian Medical University, Fuzhou 350122, China.
| | - Wei Chen
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
6
|
Lintern N, Smith AM, Jayne DG, Khaled YS. Photodynamic Stromal Depletion in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2023; 15:4135. [PMID: 37627163 PMCID: PMC10453210 DOI: 10.3390/cancers15164135] [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: 07/05/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest solid malignancies, with a five-year survival of less than 10%. The resistance of the disease and the associated lack of therapeutic response is attributed primarily to its dense, fibrotic stroma, which acts as a barrier to drug perfusion and permits tumour survival and invasion. As clinical trials of chemotherapy (CT), radiotherapy (RT), and targeted agents have not been successful, improving the survival rate in unresectable PDAC remains an urgent clinical need. Photodynamic stromal depletion (PSD) is a recent approach that uses visible or near-infrared light to destroy the desmoplastic tissue. Preclinical evidence suggests this can resensitise tumour cells to subsequent therapies whilst averting the tumorigenic effects of tumour-stromal cell interactions. So far, the pre-clinical studies have suggested that PDT can successfully mediate the destruction of various stromal elements without increasing the aggressiveness of the tumour. However, the complexity of this interplay, including the combined tumour promoting and suppressing effects, poses unknowns for the clinical application of photodynamic stromal depletion in PDAC.
Collapse
Affiliation(s)
- Nicole Lintern
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Andrew M. Smith
- Leeds Institute of Medical Research, St James’s University Hospital, Leeds LS9 7TF, UK
| | - David G. Jayne
- Leeds Institute of Medical Research, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Yazan S. Khaled
- Leeds Institute of Medical Research, St James’s University Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
7
|
Hu X, Wang G, Fang K, Li R, Dong C, Shi S, Li H. The construction of Fe-porphyrin nanozymes with peroxidase-like activity for colorimetric detection of glucose. Anal Biochem 2023:115224. [PMID: 37393976 DOI: 10.1016/j.ab.2023.115224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/20/2023] [Accepted: 06/21/2023] [Indexed: 07/04/2023]
Abstract
As a type of nanomaterials with enzyme-mimetic catalytic properties, nanozymes have attracted wide concern in biological detection. H2O2 was the characteristic product of diverse biological reactions, and the quantitative analysis for H2O2 was an important way to detect disease biomarkers, such as acetylcholine, cholesterol, uric acid and glucose. Therefore, there is of great significance for developing a simple and sensitive nanozyme to detect H2O2 and disease biomarkers by combining with corresponding enzyme. In this work, Fe-TCPP MOFs were successfully prepared by the coordination between iron ions and porphyrin ligands (TCPP). In addition, the peroxidase (POD) activity of Fe-TCPP was proved, in detail, Fe-TCPP could catalyze H2O2 to produce ·OH by Fenton reaction. Herein, glucose oxidase (GOx) was chosen as the model to build cascade reaction by combining Fe-TCPP to detect glucose. The results indicated glucose could be detected by this cascade system selectively and sensitively, and the limit of detection of glucose was achieved to 0.12 μM. Furthermore, a portable hydrogel (Fe-TCPP@GEL) was further established, which encapsulated Fe-TCPP MOFs, GOx and TMB in one system. This functional hydrogel could be applied for colorimetric detection of glucose by coupling with a smartphone easily.
Collapse
Affiliation(s)
- Xiaochun Hu
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, China; School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Guanghua Wang
- Department of Obstetrics and Gynecology, Tongren Hospital, Shanghai Jiaotong University, School of Medicine, China.
| | - Kang Fang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Ruihao Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Chunyan Dong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, China
| | - Shuo Shi
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, China.
| | - Hui Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
8
|
Hu X, Li H, Li R, Qiang S, Chen M, Shi S, Dong C. A Phase-Change Mediated Intelligent Nanoplatform for Chemo/Photothermal/Photodynamic Therapy of Cancer. Adv Healthc Mater 2023; 12:e2202245. [PMID: 36373209 DOI: 10.1002/adhm.202202245] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/23/2022] [Indexed: 11/16/2022]
Abstract
Up to now, chemotherapy is still the main strategy for cancer treatment. However, the emergence of chemo-resistance and systemic side effects often seriously affects the treatment and prognosis. Herein, an intelligent nanoplatform based on dendritic mesoporous organosilica nanoparticles (DMON) is constructed. The encapsulated phase-change material, 1-tetradecanol (TD) can serve as a "doorkeeper" and enable the responsive release of drugs based on the temperature changes. Meanwhile, polyethylene glycol (PEG) is used to improve the dispersibility and biocompatibility. Cisplatin is chosen as the model of chemotherapy drug, which is co-loaded with indocyanine green (ICG) in DMON to produce DMON-PEG-cisplatin/ICG-TD (DPCIT). Exciting, the hyperthermia and reactive oxygen species induced by ICG under the NIR-laser irradiation will initiate a phase transition of TD to release cisplatin, thus leading a combined therapy (chemo/photothermal/photodynamic therapy). The results indicated that under laser irradiation, DPCIT can kill cancer cells and inhibit tumor growth efficiently. In addition, the designed nanoplatform reveals minimal systemic toxicity in vivo, in contrast, the distinct liver damage can be observed by the direct treatment of cisplatin. Overall, this research may provide a general approach for the targeted delivery and controlled release of chemotherapy drugs to realize a cooperatively enhanced multimodal tumor therapy.
Collapse
Affiliation(s)
- Xiaochun Hu
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, P. R. China
| | - Hui Li
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, P. R. China
| | - Ruihao Li
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, P. R. China
| | - Sufeng Qiang
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, P. R. China
| | - Mengyao Chen
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, P. R. China
| | - Shuo Shi
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, P. R. China
| | - Chunyan Dong
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai, 200120, P. R. China
| |
Collapse
|
9
|
Tanaka HY, Nakazawa T, Enomoto A, Masamune A, Kano MR. Therapeutic Strategies to Overcome Fibrotic Barriers to Nanomedicine in the Pancreatic Tumor Microenvironment. Cancers (Basel) 2023; 15:cancers15030724. [PMID: 36765684 PMCID: PMC9913712 DOI: 10.3390/cancers15030724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
Pancreatic cancer is notorious for its dismal prognosis. The enhanced permeability and retention (EPR) effect theory posits that nanomedicines (therapeutics in the size range of approximately 10-200 nm) selectively accumulate in tumors. Nanomedicine has thus been suggested to be the "magic bullet"-both effective and safe-to treat pancreatic cancer. However, the densely fibrotic tumor microenvironment of pancreatic cancer impedes nanomedicine delivery. The EPR effect is thus insufficient to achieve a significant therapeutic effect. Intratumoral fibrosis is chiefly driven by aberrantly activated fibroblasts and the extracellular matrix (ECM) components secreted. Fibroblast and ECM abnormalities offer various potential targets for therapeutic intervention. In this review, we detail the diverse strategies being tested to overcome the fibrotic barriers to nanomedicine in pancreatic cancer. Strategies that target the fibrotic tissue/process are discussed first, which are followed by strategies to optimize nanomedicine design. We provide an overview of how a deeper understanding, increasingly at single-cell resolution, of fibroblast biology is revealing the complex role of the fibrotic stroma in pancreatic cancer pathogenesis and consider the therapeutic implications. Finally, we discuss critical gaps in our understanding and how we might better formulate strategies to successfully overcome the fibrotic barriers in pancreatic cancer.
Collapse
Affiliation(s)
- Hiroyoshi Y. Tanaka
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
| | - Takuya Nakazawa
- Department of Pharmaceutical Biomedicine, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
| | - Atsushi Enomoto
- Department of Pathology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya-shi 466-8550, Aichi, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai-shi 980-8574, Miyagi, Japan
| | - Mitsunobu R. Kano
- Department of Pharmaceutical Biomedicine, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
- Correspondence:
| |
Collapse
|
10
|
Zhao C, Tang X, Zhao J, Cao J, Jiang Z, Qin J. MOF derived core-shell CuO/C with temperature-controlled oxygen-vacancy for real time analysis of glucose. J Nanobiotechnology 2022; 20:507. [PMID: 36456946 PMCID: PMC9714170 DOI: 10.1186/s12951-022-01715-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022] Open
Abstract
Introducing oxygen-vacancy into the surface of the non-enzymatic sensor is supposed to be an effective way to improve inherently low catalytic activity and specificity of non-enzymatic sensors. In this work, CuO/C was synthesized at different temperatures using metal-organic frameworks as sacrificial templates to receive additional content of oxygen-vacancy. The product with the highest oxygen vacancy was found at 400 °C (named CuO/C-400 °C), which increased catalytically active sites and enhanced the charge-transfer efficiency. The sensing performance was afterward explored by amperometry under an optimal applied potential at 0.5 V (vs. SCE), presenting a broad detection range from 5.0 µM to 25.325 mM (R2 = 0.9998) with a sensitivity of 244.71 µA mM- 1 cm- 2, and a detection limit of 1 µM. Furthermore, the reliability and selectivity of CuO/C-400 °C sensors were extensively explored in the presence of artificial serum/saliva samples with gradient glucose concentrations. The human blood samples were also detected with high recoveries compared with the clinical Hexokinase method. Hence, the prepared CuO/C-400 °C sensor with a broad detection range and high selectivity can be applied for the diabetes diagnosis ex vivo without further dilution for real-time analysis in practical applications.
Collapse
Affiliation(s)
- Chen Zhao
- grid.24516.340000000123704535Shanghai Tenth People’s Hospital, School of Medicine, Tongji University Cancer Center, Tongji University, Shanghai, 200092 China ,grid.43555.320000 0000 8841 6246School of Medical Technology, School of Life Science, Beijing Institute of Technology, Beijing, 100081 China
| | - Xiaoying Tang
- grid.43555.320000 0000 8841 6246School of Medical Technology, School of Life Science, Beijing Institute of Technology, Beijing, 100081 China
| | - Jinge Zhao
- grid.43555.320000 0000 8841 6246Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081 People’s Republic of China
| | - Jie Cao
- grid.43555.320000 0000 8841 6246Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Ministry of Industry and Information Technology, Key Laboratory of Cluster Science of Ministry of Education, Beijing Key laboratory of Photoelectronic/Electro-photonic Conversion Materials, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081 People’s Republic of China
| | - Zhenqi Jiang
- grid.43555.320000 0000 8841 6246School of Medical Technology, School of Life Science, Beijing Institute of Technology, Beijing, 100081 China
| | - Jieling Qin
- grid.24516.340000000123704535Shanghai Tenth People’s Hospital, School of Medicine, Tongji University Cancer Center, Tongji University, Shanghai, 200092 China
| |
Collapse
|
11
|
Integrated energy conversion units in nanoscale frameworks induce sustained generation and amplified lethality of singlet oxygen in oxidative therapy of tumor. VIEW 2022. [DOI: 10.1002/viw.20220051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
12
|
Chen K, Sun X, Liu Y, Yang Y, Shi M, Yu J, Zhang S, Shi P. CeO 2-Decorated Metal-Organic Framework for Enhanced Photodynamic Therapy. Inorg Chem 2022; 61:16307-16316. [PMID: 36196889 DOI: 10.1021/acs.inorgchem.2c02227] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Photodynamic therapy (PDT) is quickly developing as a hopeful cancer treatment. However, hypoxic tumors, poor targeting, and photosensitizers (PS) aggregation limited the efficiency of PDT. Here, we report a hyaluronic acid (HA)-modified CeO2-nanoparticle-decorated metal-organic framework (PCN-224@CeO2-HA) to enhance PDT and achieve targeted treatment. CeO2 catalyzes H2O2 to produce O2 to solve hypoxia problems. HA could target the CD44 receptor, which is highly expressed on the tumor cell membranes. The growth of tumor cells 4T1 and MCF-7 was controlled distinctly after being incubated with PCN-224@CeO2-HA under laser irradiation, while the survival ability of normal cell LO2 was nearly unchanged. Importantly, PCN-224@CeO2-HA could be effectively aggregated within the tumor area after 12 h of injection, and the tumor growth was remarkably inhibited under laser irradiation. PCN-224@CeO2-HA presented good biocompatibility and an excellent antitumor effect, providing a new strategy to produce O2 in situ for enhanced PDT.
Collapse
Affiliation(s)
- Kaixiu Chen
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, Shandong P. R. China
| | - Xinran Sun
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, Shandong P. R. China
| | - Yingyan Liu
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, Shandong P. R. China
| | - Yapu Yang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, Shandong P. R. China
| | - Min Shi
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, Shandong P. R. China
| | - Jie Yu
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, Shandong P. R. China
| | - Shusheng Zhang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, Shandong P. R. China
| | - Pengfei Shi
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, School of Chemistry and Chemical Engineering, Linyi University, Linyi 276000, Shandong P. R. China
| |
Collapse
|
13
|
Qiang S, Hu X, Li R, Wu W, Fang K, Li H, Sun Y, Liang S, Zhao W, Wang M, Lin Y, Shi S, Dong C. CuS Nanoparticles-Loaded and Cisplatin Prodrug Conjugated Fe(III)-MOFs for MRI-Guided Combination of Chemotherapy and NIR-II Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:36503-36514. [PMID: 35925873 DOI: 10.1021/acsami.2c12727] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Ovarian cancer has become an urgent threat to global female healthcare. Cisplatin, as the traditional chemotherapeutic agent against ovarian cancer, retains several limitations, such as drug resistance and dose-limiting toxicity. In order to solve the above problems and promote the therapeutic effect of chemotherapy, combining chemotherapy and phototherapy has aroused wide interest. In this study, we constructed a versatile cisplatin prodrug-conjugated therapeutic platform based on ultrasmall CuS-modified Fe(III)-metal-organic frameworks (MIL-88) (named M-Pt/PEG-CuS) for tumor-specific enhanced synergistic chemo-/phototherapy. After intravenous injection, M-Pt/PEG-CuS presented obvious accumulation in tumor and Fe(III)-MOFs possessed magnetic resonance imaging (MRI) to guide synergy therapy. Both in vitro and in vivo experimental results showed that M-Pt/PEG-CuS could not only successfully inhibit tumor growth by combining chemotherapy and NIR-II PTT but also avoid the generation of liver damage by the direct treatment of cisplatin(II). Our work presented the development of the nanoplatform as a novel NIR-II photothermal agent, as well as gave a unique combined chemo-photothermal therapy strategy, which might provide new ways of ovarian cancer therapy for clinical translation.
Collapse
Affiliation(s)
- Sufeng Qiang
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| | - Xiaochun Hu
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| | - Ruihao Li
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| | - Wenjing Wu
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| | - Kang Fang
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| | - Hui Li
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| | - Yanting Sun
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| | - Shujing Liang
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| | - Wenrong Zhao
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| | - Mengjie Wang
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| | - Yun Lin
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| | - Shuo Shi
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| | - Chunyan Dong
- Breast Cancer Center, East Hospital Affiliated to Tongji University, Tongji University School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, People's Republic of China
| |
Collapse
|
14
|
A Fe(III)-porphyrin-oxaliplatin(IV) nanoplatform for enhanced ferroptosis and combined therapy. J Control Release 2022; 348:660-671. [PMID: 35716884 DOI: 10.1016/j.jconrel.2022.06.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/31/2022] [Accepted: 06/10/2022] [Indexed: 12/29/2022]
Abstract
Since there are several limitations in cancer treatment for traditional chemotherapy, such as side effects, poor prognosis and drug resistance, developing new combined therapy is urgently needed. In this work, a biocompatible, simple and tumor microenvironment-responsive nanotheranostics (PCN-Oxpt/PEG) was built to favor the chemotherapy/ferroptosis/immunomodulation synergism in cancer. This nanotheranostics is constructed by modifying oxaliplatin prodrug and PEG on Fe(III) - porphyrin metal-organic frameworks (PCN(Fe) MOFs). After intravenous injection, the cloak of PEG leads to long circulation, and the Fe(III)-porphyrin MOFs enables dual-model guidance with fluorescence (FL) and magnetic resonance imaging (MRI). Inside the tumor, the intracellular H2O2 would be transferred into hydroxyl radicals (•OH) by iron ions released from MOFs, which could trigger the lethal ferroptosis to cancer cells. Meanwhile, oxaliplatin(II) transformed from the loaded oxaliplatin prodrug would result in the chemotherapy, as well as immunogenic cell death (ICD), and the prodrug strategy could also avoid the occurring of liver damage by the direct administration of oxaliplatin(II). It was noticed that the ferroptosis effect was enhanced by triple-assistance during the combined therapy, as followed: (1) glutathione (GSH) would be consumed in the process of oxaliplatin(II) generation from oxaliplatin prodrug; (2) the increased CD8+ T cells induced by ICD were able to produce interferon-γ (IFN-γ), which could inhibit the transport of cystine by tumor cells, and impair the activation of glutathione peroxidase 4 (GPX4); (3) the amount of H2O2 could be increased by the internalized oxaliplatin and thus further promote the Fenton reaction and ferroptosis. Both in vivo and in vitro results revealed that tumor growing was significantly inhibited by PCN-Oxpt/PEG, taken together, the concomitant of oxaliplatin-mediated chemotherapy and ICD with triple-enhanced ferroptosis offer great prospect in the clinical treatment of cancer.
Collapse
|
15
|
Wang T, Zhang X, Xu Y, Xu Y, Zhang Y, Zhang K. Emerging nanobiotechnology-encoded relaxation tuning establishes new MRI modes to localize, monitor and predict diseases. J Mater Chem B 2022; 10:7361-7383. [PMID: 35770674 DOI: 10.1039/d2tb00600f] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Magnetic resonance imaging (MRI) is one of the most important techniques in the diagnosis of many diseases including cancers, where contrast agents (CAs) are usually necessary to improve its precision and sensitivity. Previous MRI CAs are confined to the signal-to-noise ratio (SNR) elevation of lesions for precisely localizing lesions. As nanobiotechnology advances, some new MRI CAs or nanobiotechnology-enabled MRI modes have been established to vary the longitudinal or transverse relaxation of CAs, which are harnessed to detect lesion targets, monitor disease evolution, predict or evaluate curative effect, etc. These distinct cases provide unexpected insights into the correlation of the design principles of these nanobiotechnologies and corresponding MRI CAs with their potential applications. In this review, first, we briefly present the principles, classifications and applications of conventional MRI CAs, and then elucidate the recent advances in relaxation tuning via the development of various nanobiotechnologies with emphasis on the design strategies of nanobiotechnology and the corresponding MRI CAs to target the tumor microenvironment (TME) and biological targets or activities in tumors or other diseases. In addition, we exemplified the advantages of these strategies in disease theranostics and explored their potential application fields. Finally, we analyzed the present limitations, potential solutions and future development direction of MRI after its combination with nanobiotechnology.
Collapse
Affiliation(s)
- Taixia Wang
- Central Laboratory and Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, China. .,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Xueni Zhang
- Central Laboratory and Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, China.
| | - Yuan Xu
- Central Laboratory and Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, China.
| | - Yingchun Xu
- Central Laboratory and Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, China.
| | - Yifeng Zhang
- Central Laboratory and Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, China. .,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Kun Zhang
- Central Laboratory and Ultrasound Research and Education Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, China. .,Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center for Interventional Medicine, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| |
Collapse
|
16
|
Chen C, Wu C, Yu J, Zhu X, Wu Y, Liu J, Zhang Y. Photodynamic-based combinatorial cancer therapy strategies: Tuning the properties of nanoplatform according to oncotherapy needs. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214495] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
A Review on the Efficacy and Safety of Nab-Paclitaxel with Gemcitabine in Combination with Other Therapeutic Agents as New Treatment Strategies in Pancreatic Cancer. Life (Basel) 2022; 12:life12030327. [PMID: 35330078 PMCID: PMC8953820 DOI: 10.3390/life12030327] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/10/2022] [Accepted: 02/17/2022] [Indexed: 01/27/2023] Open
Abstract
Pancreatic cancer has one of the highest mortality rates among cancers, and a combination of nab-paclitaxel with gemcitabine remains the cornerstone of first-line therapy. However, major advances are required to achieve improvements in patient outcomes. For this reason, several research groups have proposed supplementing treatment with other therapeutic agents. Ongoing studies are being conducted to find the optimal treatment in a first-line setting. In this work, we used a search strategy to compare studies on the efficacy and safety of nab-paclitaxel with gemcitabine in combination with other therapeutic agents based on the criteria of the Preferred Reporting Items for Systematic Reviews. We found seven studies in different clinical phases that met the inclusion criteria. The seven therapeutic agents were ibrutinib, necuparanib, tarextumab, apatorsen, cisplatin, enzalutamide, and momelotinib. Although these therapeutic agents have different mechanisms of action, and molecular biology studies are still needed, the present review was aimed to answer the following question: which formulations of the nab-paclitaxel/gemcitabine regimen in combination with other therapeutic agents are safest for patients with previously untreated metastatic pancreas ductal adenocarcinoma? The triple regimen is emerging as the first-line option for patients with pancreatic cancer, albeit with some limitations. Thus, further studies of this regimen are recommended.
Collapse
|
18
|
Akakuru OU, Zhang Z, Iqbal MZ, Zhu C, Zhang Y, Wu A. Chemotherapeutic nanomaterials in tumor boundary delineation: Prospects for effective tumor treatment. Acta Pharm Sin B 2022; 12:2640-2657. [PMID: 35755279 PMCID: PMC9214073 DOI: 10.1016/j.apsb.2022.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/27/2022] [Accepted: 02/06/2022] [Indexed: 12/14/2022] Open
Abstract
Accurately delineating tumor boundaries is key to predicting survival rates of cancer patients and assessing response of tumor microenvironment to various therapeutic techniques such as chemotherapy and radiotherapy. This review discusses various strategies that have been deployed to accurately delineate tumor boundaries with particular emphasis on the potential of chemotherapeutic nanomaterials in tumor boundary delineation. It also compiles the types of tumors that have been successfully delineated by currently available strategies. Finally, the challenges that still abound in accurate tumor boundary delineation are presented alongside possible perspective strategies to either ameliorate or solve the problems. It is expected that the information communicated herein will form the first compendious baseline information on tumor boundary delineation with chemotherapeutic nanomaterials and provide useful insights into future possible paths to advancing current available tumor boundary delineation approaches to achieve efficacious tumor therapy.
Collapse
Affiliation(s)
- Ozioma Udochukwu Akakuru
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China
| | - Zhoujing Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - M. Zubair Iqbal
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Chengjie Zhu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China
| | - Yewei Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, Chinese Academy of Sciences (CAS) Key Laboratory of Magnetic Materials and Devices, Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, Ningbo 315201, China
- Corresponding author.
| |
Collapse
|
19
|
Recent Advances in Strategies for Addressing Hypoxia in Tumor Photodynamic Therapy. Biomolecules 2022; 12:biom12010081. [PMID: 35053229 PMCID: PMC8774200 DOI: 10.3390/biom12010081] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 01/10/2023] Open
Abstract
Photodynamic therapy (PDT) is a treatment modality that uses light to target tumors and minimize damage to normal tissues. It offers advantages including high spatiotemporal selectivity, low side effects, and maximal preservation of tissue functions. However, the PDT efficiency is severely impeded by the hypoxic feature of tumors. Moreover, hypoxia may promote tumor metastasis and tumor resistance to multiple therapies. Therefore, addressing tumor hypoxia to improve PDT efficacy has been the focus of antitumor treatment, and research on this theme is continuously emerging. In this review, we summarize state-of-the-art advances in strategies for overcoming hypoxia in tumor PDTs, categorizing them into oxygen-independent phototherapy, oxygen-economizing PDT, and oxygen-supplementing PDT. Moreover, we highlight strategies possessing intriguing advantages such as exceedingly high PDT efficiency and high novelty, analyze the strengths and shortcomings of different methods, and envision the opportunities and challenges for future research.
Collapse
|
20
|
Zhang C, Xin L, Li J, Cao J, Sun Y, Wang X, Luo J, Zeng Y, Li Q, Zhang Y, Zhang T, Huang P. Metal-Organic Framework (MOF)-Based Ultrasound-Responsive Dual-Sonosensitizer Nanoplatform for Hypoxic Cancer Therapy. Adv Healthc Mater 2022; 11:e2101946. [PMID: 34706160 DOI: 10.1002/adhm.202101946] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/14/2021] [Indexed: 12/21/2022]
Abstract
Sonodynamic therapy (SDT), which uses reactive oxygen species to target tumors, has shown promise in the management of unresectable cancers. However, the hypoxic tumor environment limits SDT efficiency, making complete tumor destruction challenging. Here, a dual-sonosensitizer nanoplatform is developed by loading an alkyl radical generator (2,2-azobis[2-(2-imidazolin-2-yl)propane] dihydrochloride, AIPH) onto a zirconium metal-organic framework (Zr-MOF). The Zr-MOF@AIPH nanoparticles (NPs) can produce singlet oxygen, which can kill tumor cells under normoxic conditions, as well as alkyl radicals, which can kill tumor cells under both normoxic and hypoxic conditions. The combination of these free radicals further enhances SDT efficiency. Meanwhile, the nitrogen generated owing to AIPH decomposition can reduce the cavitation threshold and enhance the acoustic cavitation effect, thereby promoting NP penetration at the tumor site. Moreover, Zr-MOF@AIPH NPs exhibit good photoacoustic, fluorescence, and ultrasound imaging abilities due to their porphyrin-based structure and the nitrogen generated, which can remotely control NP delivery and determine the optimal therapeutic time window, ensuring the maximization of SDT efficiency. In vitro and in vivo examinations prove the superior antitumor efficacy, excellent biocompatibility, and favorable imaging ability of Zr-MOF@AIPH. This study spearheads the charge toward improving SDT efficacy in hypoxic environments via a combination of complementary sonosensitizers.
Collapse
Affiliation(s)
- Cong Zhang
- Department of Ultrasound in Medicine No. 88 Jiefang Road The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 P. R. China
| | - Lei Xin
- Department of Ultrasound in Medicine No. 88 Jiefang Road The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 P. R. China
| | - Jia Li
- Department of Ultrasound in Medicine No. 88 Jiefang Road The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 P. R. China
| | - Jing Cao
- Department of Ultrasound in Medicine No. 88 Jiefang Road The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 P. R. China
| | - Yu Sun
- Department of Ultrasound in Medicine No. 88 Jiefang Road The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 P. R. China
| | - Xue Wang
- Department of Ultrasound in Medicine No. 88 Jiefang Road The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 P. R. China
| | - Jiali Luo
- Department of Ultrasound in Medicine No. 88 Jiefang Road The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 P. R. China
| | - Yiqing Zeng
- Department of Ultrasound in Medicine No. 88 Jiefang Road The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 P. R. China
| | - Qunying Li
- Department of Ultrasound in Medicine No. 88 Jiefang Road The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 P. R. China
| | - Ying Zhang
- Department of Ultrasound in Medicine No. 88 Jiefang Road The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 P. R. China
| | - Tao Zhang
- Department of Ultrasound in Medicine No. 88 Jiefang Road The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 P. R. China
| | - Pintong Huang
- Department of Ultrasound in Medicine No. 88 Jiefang Road The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310009 P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering The Second Affiliated Hospital of Zhejiang University School of Medicine Zhejiang University Hangzhou 310009 P. R. China
| |
Collapse
|
21
|
Dynamic nano-assemblies based on two-dimensional inorganic nanoparticles: Construction and preclinical demonstration. Adv Drug Deliv Rev 2022; 180:114031. [PMID: 34736985 DOI: 10.1016/j.addr.2021.114031] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/07/2021] [Accepted: 10/27/2021] [Indexed: 12/14/2022]
Abstract
Dynamic drug delivery systems (DDSs) have the ability of transforming their morphology and functionality in response to the biological microenvironments at the disease site and/or external stimuli, show spatio-temporally controllable drug delivery, and enhance the treatment efficacy. Due to the large surface area and modification flexibility, two-dimensional (2D) inorganic nanomaterials are being increasingly exploited for developing intelligent DDSs for biomedical applications. In this review, we summarize the engineering methodologies used to construct transformable 2D DDSs, including changing compositions, creating defects, and surface dot-coating with polymers, biomolecules, or nanodots. Then we present and discuss dynamic inorganic 2D DDSs whose transformation is driven by the diseased characteristics, such as pH gradient, redox, hypoxia, and enzyme in the tumor microenvironment as well as the external stimuli including light, magnetism, and ultrasound. Finally, the limitations and challenges of current transformable inorganic DDSs for clinical translation and their in vivo safety assessment are discussed.
Collapse
|
22
|
Wan Y, Fu LH, Li C, Lin J, Huang P. Conquering the Hypoxia Limitation for Photodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2103978. [PMID: 34580926 DOI: 10.1002/adma.202103978] [Citation(s) in RCA: 252] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/11/2021] [Indexed: 06/13/2023]
Abstract
Photodynamic therapy (PDT) has aroused great research interest in recent years owing to its high spatiotemporal selectivity, minimal invasiveness, and low systemic toxicity. However, due to the hypoxic nature characteristic of many solid tumors, PDT is frequently limited in therapeutic effect. Moreover, the consumption of O2 during PDT may further aggravate the tumor hypoxic condition, which promotes tumor proliferation, metastasis, and invasion resulting in poor prognosis of treatment. Therefore, numerous efforts have been made to increase the O2 content in tumor with the goal of enhancing PDT efficacy. Herein, these strategies developed in past decade are comprehensively reviewed to alleviate tumor hypoxia, including 1) delivering exogenous O2 to tumor directly, 2) generating O2 in situ, 3) reducing tumor cellular O2 consumption by inhibiting respiration, 4) regulating the TME, (e.g., normalizing tumor vasculature or disrupting tumor extracellular matrix), and 5) inhibiting the hypoxia-inducible factor 1 (HIF-1) signaling pathway to relieve tumor hypoxia. Additionally, the O2 -independent Type-I PDT is also discussed as an alternative strategy. By reviewing recent progress, it is hoped that this review will provide innovative perspectives in new nanomaterials designed to combat hypoxia and avoid the associated limitation of PDT.
Collapse
Affiliation(s)
- Yilin Wan
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Lian-Hua Fu
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Chunying Li
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
23
|
Rasouli M, Fallah N, Bekeschus S. Combining Nanotechnology and Gas Plasma as an Emerging Platform for Cancer Therapy: Mechanism and Therapeutic Implication. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2990326. [PMID: 34745414 PMCID: PMC8566074 DOI: 10.1155/2021/2990326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 02/07/2023]
Abstract
Nanomedicine and plasma medicine are innovative and multidisciplinary research fields aiming to employ nanotechnology and gas plasma to improve health-related treatments. Especially cancer treatment has been in the focus of both approaches because clinical response rates with traditional methods that remain improvable for many types of tumor entities. Here, we discuss the recent progress of nanotechnology and gas plasma independently as well as in the concomitant modality of nanoplasma as multimodal platforms with unique capabilities for addressing various therapeutic issues in oncological research. The main features, delivery vehicles, and nexus between reactivity and therapeutic outcomes of nanoparticles and the processes, efficacy, and mechanisms of gas plasma are examined. Especially that the unique feature of gas plasma technology, the local and temporally controlled deposition of a plethora of reactive oxygen, and nitrogen species released simultaneously might be a suitable additive treatment to the use of systemic nanotechnology therapy approaches. Finally, we focus on the convergence of plasma and nanotechnology to provide a suitable strategy that may lead to the required therapeutic outcomes.
Collapse
Affiliation(s)
- Milad Rasouli
- Plasma Medicine Group, Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Jalale-Al-Ahmad Ave, 1411713137 Tehran, Iran
- Department of Physics and Institute for Plasma Research, Kharazmi University, 49 Dr. Mofatteh Ave, Tehran 15614, Iran
| | - Nadia Fallah
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, 49 Dr. Mofatteh Ave, 31979-37551 Tehran, Iran
| | - Sander Bekeschus
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| |
Collapse
|
24
|
Zhang T, Sun Y, Cao J, Luo J, Wang J, Jiang Z, Huang P. Intrinsic nucleus-targeted ultra-small metal-organic framework for the type I sonodynamic treatment of orthotopic pancreatic carcinoma. J Nanobiotechnology 2021; 19:315. [PMID: 34641905 PMCID: PMC8507249 DOI: 10.1186/s12951-021-01060-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/26/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Sonodynamic therapy (SDT) strategies exhibit a high tissue penetration depth and can achieve therapeutic efficacy by facilitating the intertumoral release of reactive oxygen species (ROS) with a short lifespan and limited diffusion capabilities. The majority of SDT systems developed to date are of the highly O2-dependent type II variety, limiting their therapeutic utility in pancreatic cancer and other hypoxic solid tumor types. RESULTS Herein, a nucleus-targeted ultra-small Ti-tetrakis(4-carboxyphenyl)porphyrin (TCPP) metal-organic framework (MOF) platform was synthesized and shown to be an effective mediator of SDT. This MOF was capable of generating large quantities of ROS in an oxygen-independent manner in response to low-intensity ultrasound (US) irradiation (0.5 W cm-2), thereby facilitating both type I and type II SDT. This approach thus holds great promise for the treatment of highly hypoxic orthotopic pancreatic carcinoma solid tumors. This Ti-TCPP MOF was able to induce in vitro cellular apoptosis by directly destroying DNA and inducing S phase cell cycle arrest following US irradiation. The prolonged circulation, high intratumoral accumulation, and nucleus-targeting attributes of these MOF preparations significantly also served to significantly inhibit orthotopic pancreatic tumor growth and prolong the survival of tumor-bearing mice following Ti-TCPP + US treatment. Moreover, this Ti-TCPP MOF was almost completely cleared from mice within 7 days of treatment, and no apparent treatment-associated toxicity was observed. CONCLUSION The nucleus-targeted ultra-small Ti-TCPP MOF developed herein represents an effective approach to the enhanced SDT treatment of tumors in response to low-intensity US irradiation.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District,, Hangzhou, 310009, People's Republic of China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, People's Republic of China
| | - Yu Sun
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District,, Hangzhou, 310009, People's Republic of China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, People's Republic of China
| | - Jing Cao
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District,, Hangzhou, 310009, People's Republic of China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, People's Republic of China
| | - Jiali Luo
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District,, Hangzhou, 310009, People's Republic of China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, People's Republic of China
| | - Jing Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District,, Hangzhou, 310009, People's Republic of China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, People's Republic of China
| | - Zhenqi Jiang
- Institute of Engineering Medicine, Beijing Institute of Technology, No. 5, South Street, Zhongguancun, Haidian District, Beijing, 100081, People's Republic of China.
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District,, Hangzhou, 310009, People's Republic of China.
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, People's Republic of China.
| |
Collapse
|
25
|
Ren SZ, Zhu XH, Wang B, Liu M, Li SK, Yang YS, An H, Zhu HL. A versatile nanoplatform based on multivariate porphyrinic metal-organic frameworks for catalytic cascade-enhanced photodynamic therapy. J Mater Chem B 2021; 9:4678-4689. [PMID: 34075929 DOI: 10.1039/d0tb02652b] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In recent years, the antitumor application of photodynamic therapy (PDT) has gained widespread interest in treating solid tumors. Due to the hypoxic environment in tumors, the major limit of PDT seems to be the source of oxygen. In this work, we attempted to relieve hypoxia and enhance photodynamic therapy, and therefore, designed and assembled a catalytic cascade-enhanced PDT multifunctional nanoplatform. The mentioned platform termed UIO@Ca-Pt is based on porphyrinic metal-organic framework (UIO) combination, which is simultaneously loaded by CaO2 NPs with polydopamine (PDA) and then the Pt raw material to further improve biocompatibility and efficiency. In a tumor microenvironment, CaO2 could react with water to generate calcium hydroxide and hydrogen peroxide, which was further decomposed by Pt nanoparticles to form oxygen, thereby facilitating the generation of cytotoxic singlet oxygen by photosensitizer TCPP under laser irradiation. Both in vitro and in vivo experiment results confirmed the excellent oxygen production capacity and enhanced PDT effect of UIO@Ca-Pt. With guaranteed safety in PDT, the oxygen-supplying strategy might stimulate considerable interest in the development of various metal-organic materials with multifunctionality for tumor diagnosis and therapy.
Collapse
Affiliation(s)
- Shen-Zhen Ren
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China. and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China.
| | - Xiao-Hua Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China.
| | - Bin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China.
| | - Ming Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China.
| | - Shu-Kai Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China.
| | - Yu-Shun Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China.
| | - Hailong An
- Key Laboratory of Molecular Biophysics, Hebei Province, Institute of Biophysics, School of Sciences, Hebei University of Technology, Tianjin, 300401, China.
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
26
|
Du F, Liu L, Li L, Huang J, Wang L, Tang Y, Ke B, Song L, Cheng C, Ma L, Qiu L. Conjugated Coordination Porphyrin-based Nanozymes for Photo-/Sono-Augmented Biocatalytic and Homologous Tumor Treatments. ACS APPLIED MATERIALS & INTERFACES 2021; 13:41485-41497. [PMID: 34455796 DOI: 10.1021/acsami.1c14024] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Porphyrin-based nanozymes (Porzymes) have shown promising application potential to fight against tumors using catalytically generated reactive oxygen species from the excessively produced H2O2 in the tumor microenvironment. However, the low coordination porphyrin (CP) loading ratio, difficult controllable nanostructure, low bioavailability, and low biocatalytic activities of current established Porzymes have severely limited their antitumor applications. Here, a novel malignant melanoma cell membrane-coated Pd-based CP nanoplatform (Trojan Porzymes) has been synthesized for biocatalytic and homologous tumor therapies. The Trojan Porzymes exhibit a high CP loading ratio, uniform nanoscale size, single-atom nanostructure, homologous targeted ability, and high-efficiency photo/sono-augmented biocatalytic activities. The enzyme-like biocatalytic experiments display that the Trojan Porzymes can generate abundant •OH via chemodynamic path and 1O2 via visible light or ultrasound excitation. Then we demonstrate that the Trojan Porzymes show homologous targeting ability to tumor cells and can achieve efficient accumulation and long-term retention in cancer tissues. Our in vivo data further disclose that the photo/sono-assisted chemodynamic therapies can significantly augment the treatment efficiency of malignant melanoma. We believe that our work will afford a new biocatalytic and homologous strategy for future clinical malignant melanoma treatments, which may inspire and guide more future studies to develop individualized biomedicine in precise tumor therapies.
Collapse
Affiliation(s)
- Fangxue Du
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China
| | - Luchang Liu
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China
| | - Ling Li
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China
- Department of Ultrasound, Sichuan Key Laboratory of Medical Imaging, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Jianbo Huang
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China
| | - Liyun Wang
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China
| | - Yuanjiao Tang
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China
| | - Bowen Ke
- Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Song
- Department of Pain Management, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chong Cheng
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195, Berlin, Germany
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Lang Ma
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China
| | - Li Qiu
- Department of Ultrasound, National Clinical Research Center for Geriatrics, West China Hospital, College of Polymer Science and Engineering, Sichuan University, Chengdu 610041, China
| |
Collapse
|
27
|
Karimnia V, Slack FJ, Celli JP. Photodynamic Therapy for Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13174354. [PMID: 34503165 PMCID: PMC8431269 DOI: 10.3390/cancers13174354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/16/2021] [Accepted: 08/26/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is among the most lethal of human cancers. Numerous clinical trials evaluating various combinations of chemotherapy and targeted agents and radiotherapy have failed to provide meaningful improvements in survival. A growing number of studies however have indicated that photodynamic therapy (PDT) may be a viable approach for treatment of some pancreatic tumors. PDT, which uses light to activate a photosensitizing agent in target tissue, has seen widespread adoption primarily for dermatological and other applications where superficial light delivery is relatively straightforward. Advances in fiber optic light delivery and dosimetry however have been leveraged to enable PDT even for challenging internal sites, including the pancreas. The aim of this article is to help inform future directions by reviewing relevant literature on the basic science, current clinical status, and potential challenges in the development of PDT as a treatment for PDAC. Abstract Pancreatic ductal adenocarcinoma (PDAC) is among the most lethal of human cancers. Clinical trials of various chemotherapy, radiotherapy, targeted agents and combination strategies have generally failed to provide meaningful improvement in survival for patients with unresectable disease. Photodynamic therapy (PDT) is a photochemistry-based approach that enables selective cell killing using tumor-localizing agents activated by visible or near-infrared light. In recent years, clinical studies have demonstrated the technical feasibility of PDT for patients with locally advanced PDAC while a growing body of preclinical literature has shown that PDT can overcome drug resistance and target problematic and aggressive disease. Emerging evidence also suggests the ability of PDT to target PDAC stroma, which is known to act as both a barrier to drug delivery and a tumor-promoting signaling partner. Here, we review the literature which indicates an emergent role of PDT in clinical management of PDAC, including the potential for combination with other targeted agents and RNA medicine.
Collapse
Affiliation(s)
- Vida Karimnia
- Department of Physics, University of Massachusetts at Boston, Boston, MA 02125, USA;
| | - Frank J. Slack
- Department of Pathology, BIDMC Cancer Center/Harvard Medical School, Boston, MA 02215, USA;
| | - Jonathan P. Celli
- Department of Physics, University of Massachusetts at Boston, Boston, MA 02125, USA;
- Correspondence:
| |
Collapse
|
28
|
Cao J, Sun Y, Zhang C, Wang X, Zeng Y, Zhang T, Huang P. Tablet-like TiO 2/C nanocomposites for repeated type I sonodynamic therapy of pancreatic cancer. Acta Biomater 2021; 129:269-279. [PMID: 34082101 DOI: 10.1016/j.actbio.2021.05.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022]
Abstract
Sonodynamic therapy (SDT) represents a viable approach to overcoming the limited ability of photodynamic therapy to penetrate biological barriers. However, pancreatic tumors contain a hypoxic microenvironment that limits the efficacy of oxygen-dependent type II SDT, complicating efforts to develop reliable, stable, and hypoxia-tolerant sonosensitizer. Herein, a tablet-like TiO2/C nanocomposite with a metal-organic-framework (MOF)-derived carbon structure was designed and found to be hypoxia-tolerant and stable in response to repeated ultrasound irradiation, enabling the TiO2/C-mediated generation of large quantities of reactive oxygen species (ROS) and thereby achieving efficacious type I SDT. Importantly, this nanocomposite continued to generate ROS in response to repeated ultrasound irradiation, and was able to induce tumor cell apoptosis via SDT-induced DNA damage in vitro and in vivo. This TiO2/C nanocomposite also exhibited good biocompatibility and did not induce any apparent toxicity in vitro and in vivo. Together, these data highlight TiO2/C as a valuable nanocomposite capable of facilitating repeated type I SDT, making it a promising tool for the treatment of hypoxic solid pancreatic tumors. STATEMENT OF SIGNIFICANCE: In this research, a tablet-like TiO2/C nanocomposite with a metal-organic-framework (MOF)-derived carbon structure was designed, which exhibited great stability upon repeated ultrasound irradiation, hypoxic-tolerant ability and good biocompatibility. After ultrasound irradiation, TiO2/C could efficiently generate reactive oxygen species in an oxygen-independent manner, which overcame the limitation of pure TiO2 nanoparticles. Therefore, it was applied to repeated type I sonodynamic therapy of hypoxic pancreatic tumor.
Collapse
|
29
|
Ai SL, Wang CX, Peng Y, Tu Y, Lei JJ, Xu C, Ren XH, Cheng SX. An Albumin-Based Therapeutic Nanosystem for Photosensitizer/Protein Co-Delivery to Realize Synergistic Cancer Therapy. ACS APPLIED BIO MATERIALS 2021; 4:4946-4952. [PMID: 35007043 DOI: 10.1021/acsabm.1c00233] [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] [Indexed: 12/31/2022]
Abstract
Oxygen-dependent photodynamic therapy (PDT) is hindered by the limited availability of endogenous oxygen in solid tumors and low tumor accumulation of photosensitizers. Herein, we developed a biocompatible cancer-targeted therapeutic nanosystem based on cRGD conjugated bovine serum albumin (CBSA) co-loaded with a photosensitizer (chlorin e6, Ce6) and a therapeutic protein (cytochrome c, Cytc) for synergistic photodynamic and protein therapy. The nanosystem (Ce6/Cytc@CBSA) can target αVβ3 integrin overexpressed cancer cells to improve tumor accumulation due to incorporation of cRGD. In the intracellular environment, Ce6 is released to produce toxic singlet oxygen upon near-infrared irradiation. At the same time, the therapeutic protein, Cytc, can induce programmed cell death by activating the downstream caspase pathway. Most importantly, Cytc with the catalase-like activity accelerates O2 generation by decomposing excess H2O2 in cancer cells, thereby relieving the PDT-induced hypoxia to enhance therapeutic efficacy. Both in vitro and in vivo studies reveal the significantly improved antitumor effects of the combined photodynamic/protein therapy, indicating that Ce6/Cytc@CBSA shows great potential in synergetic cancer treatments.
Collapse
Affiliation(s)
- Shu-Lun Ai
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Cai-Xia Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Yan Peng
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Yi Tu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Jin-Ju Lei
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Chang Xu
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xiao-He Ren
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| |
Collapse
|
30
|
Sun Y, Chen H, Huang Y, Xu F, Liu G, Ma L, Wang Z. One-pot synthesis of AuPd@Fe xO y nanoagent with the activable Fe species for enhanced Chemodynamic-photothermal synergetic therapy. Biomaterials 2021; 274:120821. [PMID: 33940539 DOI: 10.1016/j.biomaterials.2021.120821] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023]
Abstract
Facile fabrication of Fe-based nanotheranostic agents with the enhanced Chemodynamic therapy (CDT) effect and multiple functions is important for oncotherapy. In this report, noble-metal@FexOy core-shell nanoparticles (Au@FexOy NPs, AuRu@FexOy NPs, AuPt@FexOy NPs and AuPd@FexOy NPs) are one-pot constructed by a simply redox self-assembly strategy. As a typical example, AuPd@FexOy NPs are applied for oncotherapy. Compared to their crystalline counterparts (e.g., AuPd@c-Fe2O3 nanocrystals (NCs)), AuPd@FexOy NPs with the metastable FexOy shell can be activated by a small amount of NaBH4 to obviously enhance the production of ·OH in subsequent Fenton reaction (these activated products are termed as r-AuPd@FexOy NPs). In addition, a favorable photothermal effect (63.5% photothermal conversion efficiency) of r-AuPd@FexOy NPs can further promote the ·OH generation. Moreover, r-AuPd@FexOy NPs also show a pH-responsive T1-weighted MRI contrast property, CT imaging capacity and the function of regulating tumor microenvironment. This work presents an attractive route to prepare versatile nanotheranostic agents.
Collapse
Affiliation(s)
- Yanhong Sun
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemical Engineering, University of Science and Technology of China, Road Baohe District, Hefei, Anhui, 230026, PR China
| | - Hongda Chen
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemical Engineering, University of Science and Technology of China, Road Baohe District, Hefei, Anhui, 230026, PR China
| | - Ying Huang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemical Engineering, University of Science and Technology of China, Road Baohe District, Hefei, Anhui, 230026, PR China
| | - Fengqin Xu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemical Engineering, University of Science and Technology of China, Road Baohe District, Hefei, Anhui, 230026, PR China
| | - Guifeng Liu
- Department of Radiology, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, 130033, PR China.
| | - Lina Ma
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.
| | - Zhenxin Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemical Engineering, University of Science and Technology of China, Road Baohe District, Hefei, Anhui, 230026, PR China.
| |
Collapse
|
31
|
Li A, Yang X, Chen J. A novel route to size-controlled MIL-53(Fe) metal-organic frameworks for combined chemodynamic therapy and chemotherapy for cancer. RSC Adv 2021; 11:10540-10547. [PMID: 35423581 PMCID: PMC8695691 DOI: 10.1039/d0ra09915e] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/19/2021] [Indexed: 12/31/2022] Open
Abstract
Metal-organic frameworks (MOFs), such as MIL-53(Fe), have considerable potential as drug carriers in cancer treatment due to their notable characteristics, including controllable particle sizes, high catalytic activity, biocompatibility and large porosity, and are widely used in a broad range of drugs. In this study, a new approach for the synthesis of MIL-53(Fe) nanocrystals with controlled sizes has been developed using a non-ionic surfactant PVP as the conditioning and stabilizing agent, respectively. During the nucleation of MIL-53(Fe), the PVP droplet, as a nano-reactor, controlled the growth of the crystal nucleus. The size and aspect ratio (length/width) of nanocrystals increased with an increase in PVP in the synthetic mixture. The MIL-53(Fe) nanocrystals showed a homogeneous morphology, with approximately 190 nm in length and 100 nm in width. MIL-53(Fe) not only was used to load the anticancer drug doxorubicin (DOX) but also generated hydroxyl radicals (˙OH) via a Fenton-like reaction for ROS-mediated/chemo-therapy of cancer cells. The approach was expected to synthesize numerous types of nano-size iron(iii)-based MOFs, such as MIL-53, 89, 88A, 88B and 101. The MIL-53(Fe) nanocrystals hold great promise as a candidate to improve the controlled release of drugs and treatment effect for cancer therapy.
Collapse
Affiliation(s)
- Anxia Li
- Sanya Central Hospital (Hainan Third People's Hospital) Sanya Hainan 572000 China
| | - Xiaoxin Yang
- College of Chemistry and Chemical Engineering, Hunan University Changsha Hunan 410082 China
| | - Juan Chen
- Sanya Central Hospital (Hainan Third People's Hospital) Sanya Hainan 572000 China
| |
Collapse
|
32
|
Liu X, Liang T, Zhang R, Ding Q, Wu S, Li C, Lin Y, Ye Y, Zhong Z, Zhou M. Iron-Based Metal-Organic Frameworks in Drug Delivery and Biomedicine. ACS APPLIED MATERIALS & INTERFACES 2021; 13:9643-9655. [PMID: 33606494 DOI: 10.1021/acsami.0c21486] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Metal-organic frameworks (MOFs) are crystalline materials comprising metal centers and organic linkers that feature structural rigidity and functional flexibility. These attractive materials offer large surface areas, high porosity, and good chemical stability; they have shown promise in chemistry (H2 separation and catalysis), magnetism, and optics. They have also shown potential for drug delivery following the demonstration in 2006 that chromium-based MOFs can be loaded with ibuprofen. Since then, iron-based MOFs (Fe-MOFs) have been shown to offer high drug loading and excellent biocompatibility. The present review focuses on the synthesis and surface modifications of Fe-MOFs as well as their applications in drug delivery and biomedicine.
Collapse
Affiliation(s)
- Xianbin Liu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Tiantian Liang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Rongtao Zhang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qian Ding
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Siqiong Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Lin
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yun Ye
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhirong Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
33
|
Zhou H, Qin F, Chen C. Designing Hypoxia-Responsive Nanotheranostic Agents for Tumor Imaging and Therapy. Adv Healthc Mater 2021; 10:e2001277. [PMID: 32985141 DOI: 10.1002/adhm.202001277] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/06/2020] [Indexed: 12/15/2022]
Abstract
Hypoxia, a common feature of most solid tumors, plays an important role in tumor proliferation, metastasis, and invasion, leading to drug, radiation, and photodynamic therapy resistance, and resulting in a sharp reduction in the disease-free survival rate of tumor patients. The lack of sufficient blood supply to the interior regions of tumors hinders the delivery of traditional drugs and contrast agents, interfering with their accumulation in the hypoxic region, and preventing efficient theranostics. Thus, there is a need for the fabrication of novel tumor theranostic agents that overcome these obstacles. Reports, in recent years, of hypoxia-responsive nanomaterials may provide with such means. In this review, a comprehensive description of the physicochemical and biological characteristics of hypoxic tumor tissues is provided, the principles of designing the hypoxia-responsive tumor theranostic agents are discussed, and the recent research into hypoxia-triggered nanomaterials is examined. Additionally, other hypoxia-associated responsive strategies, the current limitations, and future prospects for hypoxia-responsive nanotheranostic agents in tumor treatment are discussed.
Collapse
Affiliation(s)
- Huige Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology (NCNST) Beijing 100190 China
- College of Materials Sciences and Opto‐Electronic Technology University of Chinese Academy of Sciences Beijing 100049 China
- Research Unit of Nanoscience and Technology Chinese Academy of Medical Sciences Beijing 100190 China
| | - Fenglan Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology (NCNST) Beijing 100190 China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology (NCNST) Beijing 100190 China
- College of Materials Sciences and Opto‐Electronic Technology University of Chinese Academy of Sciences Beijing 100049 China
- Research Unit of Nanoscience and Technology Chinese Academy of Medical Sciences Beijing 100190 China
| |
Collapse
|
34
|
Yang H, Liu R, Xu Y, Qian L, Dai Z. Photosensitizer Nanoparticles Boost Photodynamic Therapy for Pancreatic Cancer Treatment. NANO-MICRO LETTERS 2021; 13:35. [PMID: 34138222 PMCID: PMC8187547 DOI: 10.1007/s40820-020-00561-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 10/31/2020] [Indexed: 05/13/2023]
Abstract
Patients with pancreatic cancer (PCa) have a poor prognosis apart from the few suitable for surgery. Photodynamic therapy (PDT) is a minimally invasive treatment modality whose efficacy and safety in treating unresectable localized PCa have been corroborated in clinic. Yet, it suffers from certain limitations during clinical exploitation, including insufficient photosensitizers (PSs) delivery, tumor-oxygenation dependency, and treatment escape of aggressive tumors. To overcome these obstacles, an increasing number of researchers are currently on a quest to develop photosensitizer nanoparticles (NPs) by the use of a variety of nanocarrier systems to improve cellular uptake and biodistribution of photosensitizers. Encapsulation of PSs with NPs endows them significantly higher accumulation within PCa tumors due to the increased solubility and stability in blood circulation. A number of approaches have been explored to produce NPs co-delivering multi-agents affording PDT-based synergistic therapies for improved response rates and durability of response after treatment. This review provides an overview of available data regarding the design, methodology, and oncological outcome of the innovative NPs-based PDT of PCa.
Collapse
Affiliation(s)
- Huanyu Yang
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing, 100050, People's Republic of China
| | - Renfa Liu
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, People's Republic of China
| | - Yunxue Xu
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, People's Republic of China
| | - Linxue Qian
- Department of Ultrasound, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing, 100050, People's Republic of China.
| | - Zhifei Dai
- Department of Biomedical Engineering, College of Engineering, Peking University, No. 5 Yiheyuan Road, Haidian District, Beijing, 100871, People's Republic of China.
| |
Collapse
|
35
|
Wang B, Dai Y, Kong Y, Du W, Ni H, Zhao H, Sun Z, Shen Q, Li M, Fan Q. Tumor Microenvironment-Responsive Fe(III)-Porphyrin Nanotheranostics for Tumor Imaging and Targeted Chemodynamic-Photodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:53634-53645. [PMID: 33205657 DOI: 10.1021/acsami.0c14046] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The development of effective and safe tumor nanotheranostics remains a research imperative. Herein, tumor microenvironment (TME)-responsive Fe(III)-porphyrin (TCPP) coordination nanoparticles (FT@HA NPs) were prepared using a simple one-pot method followed by modification with hyaluronic acid (HA). FT@HA NPs specifically accumulated in CD44 receptor-overexpressed tumor tissues through the targeting property of HA and upon endocytosis by tumor cells. After cell internalization, intracellular acidic microenvironments and high levels of glutathione (GSH) triggered the rapid decomposition of FT@HA NPs to release free TCPP molecules and Fe(III) ions. The released Fe(III) ions could trigger GSH depletion and Fenton reaction, activating chemodynamic therapy (CDT). Meanwhile, the fluorescence and photodynamic effects of the TCPP could be also activated, achieving controlled reactive oxygen species (ROS) generation and avoiding side effects on normal tissues. Moreover, the rapid consumption of GSH further enhanced the efficacy of CDT and photodynamic therapy (PDT). The in vivo experiments further demonstrated that the antitumor effect of these nanotheranostics was significantly enhanced and that their toxicity and side effects against normal tissues were effectively suppressed. The FT@HA NPs can be applied for activated tumor combination therapy under the guidance of dual-mode imaging including fluorescence imaging and magnetic resonance imaging, providing an effective strategy for the design and preparation of TME-responsive multifunctional nanotheranostics for precise tumor imaging and combination therapy.
Collapse
Affiliation(s)
- Bing Wang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Yeneng Dai
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Yingjie Kong
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Wenyu Du
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Haiyang Ni
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Honghai Zhao
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Zhiquan Sun
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Qingming Shen
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Meixing Li
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Quli Fan
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| |
Collapse
|
36
|
Sundaram A, Peng L, Chai L, Xie Z, Ponraj JS, Wang X, Wang G, Zhang B, Nie G, Xie N, Rajesh Kumar M, Zhang H. Advanced nanomaterials for hypoxia tumor therapy: challenges and solutions. NANOSCALE 2020; 12:21497-21518. [PMID: 33094770 DOI: 10.1039/d0nr06271e] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
In recent years, nanomaterials and nanotechnology have emerged as vital factors in the medical field with a unique contribution to cancer medicine. Given the increasing number of cancer patients, it is necessarily required to develop innovative strategies and therapeutic modalities to tackle hypoxia, which forms a hallmark and great barrier in treating solid tumors. The present review details the challenges in nanotechnology-based hypoxia, targeting the strategies and solutions for better therapeutic performances. The interaction between hypoxia and tumor is firstly introduced. Then, we review the recently developed engineered nanomaterials towards multimodal hypoxia tumor therapies, including chemotherapy, radiotherapy, and sonodynamic treatment. In the next part, we summarize the nanotechnology-based strategies for overcoming hypoxia problems. Finally, current challenges and future directions are proposed for successfully overcoming the hypoxia tumor problems.
Collapse
Affiliation(s)
- Aravindkumar Sundaram
- Department of Orthopaedic Surgery, the Sixth Affiliated Hospital of Guangzhou Medical University, 511508 Qingyuan, Guangdong, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Alsaab HO, Alghamdi MS, Alotaibi AS, Alzhrani R, Alwuthaynani F, Althobaiti YS, Almalki AH, Sau S, Iyer AK. Progress in Clinical Trials of Photodynamic Therapy for Solid Tumors and the Role of Nanomedicine. Cancers (Basel) 2020; 12:E2793. [PMID: 33003374 PMCID: PMC7601252 DOI: 10.3390/cancers12102793] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 01/03/2023] Open
Abstract
Current research to find effective anticancer treatments is being performed on photodynamic therapy (PDT) with increasing attention. PDT is a very promising therapeutic way to combine a photosensitive drug with visible light to manage different intense malignancies. PDT has several benefits, including better safety and lower toxicity in the treatment of malignant tumors over traditional cancer therapy. This reasonably simple approach utilizes three integral elements: a photosensitizer (PS), a source of light, and oxygen. Upon light irradiation of a particular wavelength, the PS generates reactive oxygen species (ROS), beginning a cascade of cellular death transformations. The positive therapeutic impact of PDT may be limited because several factors of this therapy include low solubilities of PSs, restricting their effective administration, blood circulation, and poor tumor specificity. Therefore, utilizing nanocarrier systems that modulate PS pharmacokinetics (PK) and pharmacodynamics (PD) is a promising approach to bypassing these challenges. In the present paper, we review the latest clinical studies and preclinical in vivo studies on the use of PDT and progress made in the use of nanotherapeutics as delivery tools for PSs to improve their cancer cellular uptake and their toxic properties and, therefore, the therapeutic impact of PDT. We also discuss the effects that photoimmunotherapy (PIT) might have on solid tumor therapeutic strategies.
Collapse
Affiliation(s)
- Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia;
| | - Maha S. Alghamdi
- Department of Pharmaceutical Care, King Abdul-Aziz Specialist Hospital (KAASH), Taif 26521, Saudi Arabia;
| | - Albatool S. Alotaibi
- College of Pharmacy, Taif University, Al Haweiah, Taif 21944, Saudi Arabia; (A.S.A.); (F.A.)
| | - Rami Alzhrani
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia;
| | - Fatimah Alwuthaynani
- College of Pharmacy, Taif University, Al Haweiah, Taif 21944, Saudi Arabia; (A.S.A.); (F.A.)
| | - Yusuf S. Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia;
| | - Atiah H. Almalki
- Department of Pharmaceutical chemistry, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia;
| | - Samaresh Sau
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48021, USA; (S.S.); (A.K.I.)
| | - Arun K. Iyer
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48021, USA; (S.S.); (A.K.I.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|