1
|
Yu X, Zhang Q, Wang L, Zhang Y, Zhu L. Engineered nanoparticles for imaging and targeted drug delivery in hepatocellular carcinoma. Exp Hematol Oncol 2025; 14:62. [PMID: 40307921 PMCID: PMC12044934 DOI: 10.1186/s40164-025-00658-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/18/2025] [Indexed: 05/02/2025] Open
Abstract
Liver cancer, notably hepatocellular carcinoma (HCC), poses a significant global health burden due to its high fatality rates. Conventional antitumor medications face challenges, including poor targeting, high toxicity, and drug resistance, leading to suboptimal clinical outcomes. This review focused on nanoparticle use in diagnosing and delivering medication for HCC, aiming to advance the development of nanomedicines for improved treatment outcomes. As an emerging frontier science and technology, nanotechnology has shown great potential, especially in precision medicine and personalized treatment. The success of nanosystems is attributable to their smaller size, biocompatibility, selective tumor accumulation, and lower toxicity. Nanoparticles, as a central part of nanotechnology innovation, have emerged in the field of medical diagnostics and therapeutics to overcome the various limitations of conventional chemotherapy, thus offering promising applications for improved selectivity, earlier and more precise diagnosis of cancers, personalized treatment, and overcoming drug resistance. Nanoparticles play a crucial role in drug delivery and imaging of HCC, with the body acting as a delivery system to target and deliver drugs or diagnostic reagents to specific organs or tissues, helping to accurately diagnose and target therapies while minimizing damage to healthy tissues. They protect drugs from early degradation and increase their biological half-life.
Collapse
Affiliation(s)
- Xianzhe Yu
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, No. 10 Qinyun Nan Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Qin Zhang
- Department of Postgraduate Students, West China School of Medicine/West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Leibo Wang
- Department of Surgery, Beijing Jishuitan Hospital Guizhou Hospital Guiyang, Guiyang, 550000, Guizhou, The People's Republic of China
| | - Yan Zhang
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Lingling Zhu
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
2
|
Cai L, Du Y, Xiong H, Zheng H. Application of nanotechnology in the treatment of hepatocellular carcinoma. Front Pharmacol 2024; 15:1438819. [PMID: 39679376 PMCID: PMC11637861 DOI: 10.3389/fphar.2024.1438819] [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: 05/26/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
Hepatocellular carcinoma is the predominant histologic variant of hepatic malignancy and has become a major challenge to global health. The increasing incidence and mortality of hepatocellular carcinoma has created an urgent need for effective prevention, diagnosis, and treatment strategies. This is despite the impressive results of multiple treatments in the clinic. However, the unique tumor immunosuppressive microenvironment of hepatocellular carcinoma increases the difficulty of treatment and immune tolerance. In recent years, the application of nanoparticles in the treatment of hepatocellular carcinoma has brought new hope for tumor patients. Nano agents target tumor-associated fibroblasts, regulatory T cells, myeloid suppressor cells, tumor-associated macrophages, tumor-associated neutrophils, and immature dendritic cells, reversed the immunosuppressive microenvironment of hepatocellular carcinoma. In addition, he purpose of this review is to summarize the advantages of nanotechnology in guiding surgical excision, local ablation, TACE, standard chemotherapy, and immunotherapy, application of nano-vaccines has also continuously enriched the treatment of liver cancer. This study aims to investigate the potential applications of nanotechnology in the management of hepatocellular carcinoma, with the ultimate goal of enhancing therapeutic outcomes and improving the prognosis for patients affected by this malignancy.
Collapse
Affiliation(s)
| | | | | | - Honggang Zheng
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Bhange M, Telange DR. Unlocking the Potential of Phyto Nanotherapeutics in Hepatocellular Carcinoma Treatment: A Review. J Hepatocell Carcinoma 2024; 11:2241-2256. [PMID: 39574434 PMCID: PMC11579138 DOI: 10.2147/jhc.s483619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/03/2024] [Indexed: 11/24/2024] Open
Abstract
Hepatocellular carcinoma is the fifth leading cancer in related diseases most commonly in men and women. The curative treatments of liver cancer are short-listed, associated with toxicities and therapeutically. Emerging nanotechnologies exhibited the possibility to treat or target liver cancer. Over the years, to phytosome solid lipid nanoparticles, gold, silver, liposomes, and phospholipid nanoparticles have been produced for liver cancer therapy, and some evidence of their effectiveness has been established. Ideas are limited to the laboratory scale, and in order to develop active targeting of nanomedicine for the clinical aspects, they must be extended to a larger scale. Thus, the current review focuses on previously and presently published research on the creation of phytosomal nanocarriers for the treatment of hepatocellular carcinoma. In hepatocellular carcinoma (HCC), phytosomal nanotherapeutics improve the targeted delivery and bioavailability of phytochemicals to tumor cells, thereby reducing systemic toxicity and increasing therapeutic efficacy. In order to address the intricate molecular processes implicated in HCC, this strategy is essential.
Collapse
Affiliation(s)
- Manjusha Bhange
- Department of Pharmaceutics, Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education & Research (DU), Wardha, Maharashtra, India
| | - Darshan R Telange
- Department of Pharmaceutics, Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education & Research (DU), Wardha, Maharashtra, India
| |
Collapse
|
4
|
Liu Y, Wu Y, Li Z, Wan D, Pan J. Targeted Drug Delivery Strategies for the Treatment of Hepatocellular Carcinoma. Molecules 2024; 29:4405. [PMID: 39339402 PMCID: PMC11434448 DOI: 10.3390/molecules29184405] [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: 08/21/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent malignant tumors, exhibiting a high incidence rate that presents a substantial threat to human health. The use of sorafenib and lenvatinib, commonly employed as single-agent targeted inhibitors, complicates the treatment process due to the absence of definitive targeting. Nevertheless, the advent of nanotechnology has injected new optimism into the domain of liver cancer therapy. Nanocarriers equipped with active targeting or passive targeting mechanisms have demonstrated the capability to deliver drugs to tumor cells with high efficiency. This approach not only facilitates precise delivery to the affected site but also enables targeted drug release, thereby enhancing therapeutic efficacy. As medical technology progresses, there is an increasing call for innovative treatment modalities, including novel chemotherapeutic agents, gene therapy, phototherapy, immunotherapy, and combinatorial treatments for HCC. These emerging therapies are anticipated to yield improved clinical outcomes for patients, while minimizing systemic toxicity and adverse effects. Consequently, the application of nanotechnology is poised to significantly improve HCC treatment. This review focused on targeted strategies for HCC and the application of nanotechnology in this area.
Collapse
Affiliation(s)
- Yonghui Liu
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| | - Yanan Wu
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| | - Zijian Li
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Dong Wan
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
- School of Chemical Engineering and Technology, Tiangong University, Tianjin 300387, China
| | - Jie Pan
- School of Chemistry, Tiangong University, Tianjin 300387, China; (Y.L.)
| |
Collapse
|
5
|
Su Y, Huang L, Zhang D, Zeng Z, Hong S, Lin X. Recent Advancements in Ultrasound Contrast Agents Based on Nanomaterials for Imaging. ACS Biomater Sci Eng 2024; 10:5496-5512. [PMID: 39246058 DOI: 10.1021/acsbiomaterials.4c00890] [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] [Indexed: 09/10/2024]
Abstract
Ultrasound (US) is a type of mechanical wave that is capable of transmitting energy through biological tissues. By utilization of various frequencies and intensities, it can elicit specific biological effects. US imaging (USI) technology has been continuously developed with the advantages of safety and the absence of radiation. The advancement of nanotechnology has led to the utilization of various nanomaterials composed of both organic and inorganic compounds as ultrasound contrast agents (UCAs). These UCAs enhance USI, enabling real-time monitoring, diagnosis, and treatment of diseases, thereby facilitating the widespread adoption of UCAs in precision medicine. In this review, we introduce various UCAs based on nanomaterials for USI. Their principles can be roughly divided into the following categories: carrying and transporting gases, endogenous gas production, and the structural characteristics of the nanomaterial itself. Furthermore, the synergistic benefits of US in conjunction with various imaging modalities and their combined application in disease monitoring and diagnosis are introduced. In addition, the challenges and prospects for the development of UCAs are also discussed.
Collapse
Affiliation(s)
- Yina Su
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Linjie Huang
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Dongdong Zhang
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Zheng Zeng
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Shanni Hong
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| | - Xiahui Lin
- School of Medical Imaging, Fujian Medical University, Fuzhou 350122, Fujian, P. R. China
| |
Collapse
|
6
|
Metkar SP, Fernandes G, Navti PD, Nikam AN, Kudarha R, Dhas N, Seetharam RN, Santhosh KV, Rao BSS, Mutalik S. Nanoparticle drug delivery systems in hepatocellular carcinoma: A focus on targeting strategies and therapeutic applications. OPENNANO 2023; 12:100159. [DOI: 10.1016/j.onano.2023.100159] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
7
|
Ling J, Jiang Y, Yan S, Dang H, Yue H, Liu K, Kuang L, Liu X, Tang H. A novel pH- and glutathione-responsive drug delivery system based on in situ growth of MOF199 on mesoporous organic silica nanoparticles targeting the hepatocellular carcinoma niche. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00139-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
For people with advanced hepatocellular carcinoma (HCC), systemic chemotherapy remains the only choice of palliative treatment. However, chemotherapy efficacy is not effective due to its short blood circulation times, nonspecific cell and tissue biodistribution, and rapid metabolism or excretion from the body. Therefore, a targeted nanomedicine delivery system is urgently needed.
Methods
In order to improve the treatment efficiency of HCC, based on in situ growth of a copper metal organic framework on mesoporous organic silica nanoparticles, dual pH- and glutathione (GSH)-responsive multifunctional nanocomposites were synthesized as nanocarriers for enhanced HCC therapy. In this research, cellular uptake studies were performed using CLSM and Bio-TEM observations. Flow cytometry, AO-EB fluorescent staining, EdU test and Western blot were utilized to explore the apoptosis and proliferation process. In vivo imaging was employed to research the distribution of the nanocomposites in HCC tumor-bearing nude mice and the xenograft model of HCC tumor-bearing nude mice was applied to investigate the anti-tumor effects of drug-loaded nanocomposites in vivo.
Results
This newly constructed degradable nanocomposite DOX/SOR@SP94 and mPEG-anchored MONs@MOF199 (D/S@SPMM) has the benefits of controllable pore size, high encapsulation efficiency, and precise targeting. According to the results of in vivo imaging and anti-tumor studies, as well as pharmacokinetic research, D/S@SPMM possessed precise HCC tumor targeting and long-lasting accumulation properties at the tumor region. Compared with traditional chemotherapy and non-targeted drug delivery systems, anti-tumor efficiency was increased by approximately 10- and 5-fold, respectively. The nanocomposites exhibited excellent anti-tumor properties without inducing observable systemic toxicity, owing to efficient DOX and SOR loading and release as well as the HCC specific targeting peptide SP94.
Conclusions
The in vitro and in vivo anti-tumor results indicated that these nanocomposites could be an efficient nanomedicine for targeting HCC therapy.
Collapse
|
8
|
Wu D, Li Y, Zhu L, Zhang W, Xu S, Yang Y, Yan Q, Yang G. A biocompatible superparamagnetic chitosan-based nanoplatform enabling targeted SN-38 delivery for colorectal cancer therapy. Carbohydr Polym 2021; 274:118641. [PMID: 34702462 DOI: 10.1016/j.carbpol.2021.118641] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 08/12/2021] [Accepted: 09/01/2021] [Indexed: 11/26/2022]
Abstract
7-Ethyl-10-hydroxycamptothecin (SN-38) as a potent anti-tumor candidate, suffers the constraints from its poor water solubility, pH-dependent lactone ring stability and the lack of efficient delivery system without losing its activity. Herein, biocompatible superparamagnetic chitosan-based nanocomplexes complexing with water-soluble polymeric prodrug poly(L-glutamic acid)-SN-38 (PGA-SN-38) was engineered for efficient delivery of SN-38. The manufacturing process of colloidal complexes was green, expeditious and facile, with one-shot addition of PGA-SN-38 into chitosan solution without using any organic solvent or surfactant. Upon introducing ultra-small-size superparamagnetic nanoparticles (~10 nm), the developed magnetic nanocomplexes exhibited significantly boosted tumor-targeted accumulation and efficient cellular internalization under a local magnetic field. Notably, the magnetic nanocomplexes achieved distinctly superior targeting and anti-tumor efficacy in the established xenograft colorectal cancer model of mice, with high tumor suppression rate up to 81%. Therefore, this superparamagnetic chitosan-based nanocomplex system could provide a promising platform for the targeted delivery of SN-38 in colorectal cancer therapy.
Collapse
Affiliation(s)
- Danjun Wu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yi Li
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Lixi Zhu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wangyang Zhang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Shumin Xu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yan Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Qinying Yan
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Gensheng Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
9
|
Mintz KJ, Leblanc RM. The use of nanotechnology to combat liver cancer: Progress and perspectives. Biochim Biophys Acta Rev Cancer 2021; 1876:188621. [PMID: 34454983 DOI: 10.1016/j.bbcan.2021.188621] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 01/04/2023]
Abstract
Liver cancer is one of the most common cancers worldwide and is also one of the most difficult cancers to treat, resulting in almost one million deaths per year, and the danger of this cancer is compounded when the tumor is nonresectable. Hepatocellular carcinoma (HCC) is the most common type of liver cancer and has the third highest mortality rate worldwide. Considering the morbid statistics surrounding this cancer it is a popular research topic to target for better therapy practices. This review summarizes the role of nanotechnology in these endeavors. Nanoparticles (NPs) are a very broad class of material and many different kinds have been used to potentially combat liver cancer. Gold, silver, platinum, metal oxide, calcium, and selenium NPs as well as less common materials are all inorganic NPs that have been used as a therapeutic, carrier, or imaging agent in drug delivery systems (DDS) and these efforts are described. Carbon-based NPs, including polymeric, polysaccharide, and lipid NPs as well as carbon dots, have also been widely studied for this purpose and the role they play in DDS for the treatment of liver cancer is illustrated in this review. The multifunctional nature of many NPs described herein, allows these systems to display high anticancer activity in vitro and in vivo and highlights the advantage of and need for combinatorial therapy in treating this difficult cancer. These works are summarized, and future directions are presented for this promising field.
Collapse
Affiliation(s)
- Keenan J Mintz
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA; Department of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Roger M Leblanc
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA.
| |
Collapse
|
10
|
Zeng F, Peng K, Han L, Yang J. Photothermal and Photodynamic Therapies via NIR-Activated Nanoagents in Combating Alzheimer's Disease. ACS Biomater Sci Eng 2021; 7:3573-3585. [PMID: 34279071 DOI: 10.1021/acsbiomaterials.1c00605] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
It is well established that the polymerization of amyloid-β peptides into fibrils/plaques is a critical step during the development of Alzheimer's disease (AD). Phototherapy, which includes photodynamic therapy and photothermal therapy, is a highly attractive strategy in AD treatment due to its merits of operational flexibility, noninvasiveness, and high spatiotemporal resolution. Distinct from traditional chemotherapies or immunotherapies, phototherapies capitalize on the interaction between photosensitizers or photothermal transduction agents and light to trigger photochemical reactions to generate either reactive oxygen species or heat effects to modulate Aβ aggregation, ultimately restoring nerve damage and ameliorating memory deficits. In this Review, we provide an overview of the recent advances in the development of near-infrared-activated nanoagents for AD phototherapies and discuss the potential challenges of and perspectives on this emerging field with a special focus on how to improve the efficiency and utility of such treatment. We hope that this Review will spur preclinical research and the clinical translation of AD treatment through phototherapy.
Collapse
Affiliation(s)
- Fantian Zeng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Kewen Peng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Ling Han
- Center for Drug Evaluation, National Medical Products Administration, Beijing 100022, China
| | - Jian Yang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| |
Collapse
|
11
|
Li S, Yang S, Liu C, He J, Li T, Fu C, Meng X, Shao H. Enhanced Photothermal-Photodynamic Therapy by Indocyanine Green and Curcumin-Loaded Layered MoS 2 Hollow Spheres via Inhibition of P-Glycoprotein. Int J Nanomedicine 2021; 16:433-442. [PMID: 33488079 PMCID: PMC7815073 DOI: 10.2147/ijn.s275938] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/30/2020] [Indexed: 12/30/2022] Open
Abstract
PURPOSE P-glycoprotein (P-gp), which is highly expressed in liver cancer cells, is one of the obstacles for the treatment of cancer. In this study, we have prepared and characterized a kind of novel ICG&Cur@MoS2 (ICG and Cur represent indocyanine green and curcumin, respectively) nanoplatform, which can achieve photothermal-photodynamic therapy and inhibit the P-gp effectively and safely. METHODS In this work, plenty of studies including drug release, acute toxicity, Western blot, real-time PCR, cell viability, therapeutic experiment in vivo, immunofluorescence and so on were conducted to test the antitumor potential of ICG&Cur@MoS2 and the inhibitory effect of curcumin on P-gp. RESULTS The ICG&Cur@MoS2 NPs exhibit an excellent photothermal effect and relatively low toxicity. Cell viability in the ICG&Cur@MoS2 + NIR group was significantly lower than that in ICG@MoS2 + NIR group (75.3% vs 81.2%, 59.0% vs 64.4%, 20.3% vs 27.5%, and 15.4% vs 22.3%) at the concentration of ICG at 0.5, 5, 25, 50 μg/mL (P<0.05 at each concentration). Western blot, Q-PCR, and immunofluorescence assay indicate ICG&Cur@MoS2 NPs can inhibit the P-gp effectively and safely. In vivo, the tumors in the ICG@MoS2 + NIR group are significantly smaller than those in the MoS2 + NIR group (95.0 vs 420.9 mm3, p<0.05). CONCLUSION In conclusion, we have successfully synthesized ICG&Cur@MoS2 nanoparticles which can not only achieve PTT-PDT but also inhibit P-gp effectively. Our findings indicate that the PTT-PDT exhibits great potential in the treatment of hepatocellular carcinoma. Meanwhile, ICG&Cur@MoS2 can effectively inhibit the expression of P-gp, which will enhance the PDT effect.
Collapse
Affiliation(s)
- Shuai Li
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang110001, Liaoning, People’s Republic of China
| | - Shuping Yang
- Department of Pain Medicine, The First Hospital of China Medical University, Shenyang110001, Liaoning, People’s Republic of China
| | - Chong Liu
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang110001, Liaoning, People’s Republic of China
| | - Jintong He
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang110001, Liaoning, People’s Republic of China
| | - Tian Li
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang110001, Liaoning, People’s Republic of China
| | - Changhui Fu
- Laboratory of Controllable Preparation and Application of Nanomaterials, Chinese Academy of Sciences Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing100190, People’s Republic of China
| | - Xianwei Meng
- Laboratory of Controllable Preparation and Application of Nanomaterials, Chinese Academy of Sciences Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing100190, People’s Republic of China
| | - Haibo Shao
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang110001, Liaoning, People’s Republic of China
| |
Collapse
|
12
|
Huang M, Zhao Q, Ye Z, Xu D, Tang S, Jiang T. Development of a novel melatonin-modified near-infrared fluorescent probe for in vivo hepatocellular carcinoma imaging. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2020; 12:4556-4561. [PMID: 33001063 DOI: 10.1039/d0ay01135e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy worldwide with poor prognosis. The early identification and precise resection of HCC are essential for improving the prognosis and overall survival of patients. In clinical practice, fluorescence imaging is a powerful technology to identify and remove HCC lesions, but accurate and reliable detection of HCC continues to remain a challenge due to non-specificity and false-positive uptake of probes. To circumvent these problems, it is crucial to design a specific probe for the accurate detection of HCC. Herein, we reported the design and synthesis of an NIR fluorescent probe by conjugating IRDye800CW with melatonin, which plays a significant role in the HCC development. The in vivo imaging revealed that IRDye800-MT was uptake specifically by the HCC tumor with a high tumor-to-background ratio. These results demonstrated that IRDye800-MT might hold clinical potentials for future diagnosis of HCC patients.
Collapse
Affiliation(s)
- Min Huang
- Department of Ultrasound, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310003, China.
| | | | | | | | | | | |
Collapse
|
13
|
Zhang F, Liu S, Zhang N, Kuang Y, Li W, Gai S, He F, Gulzar A, Yang P. X-ray-triggered NO-released Bi-SNO nanoparticles: all-in-one nano-radiosensitizer with photothermal/gas therapy for enhanced radiotherapy. NANOSCALE 2020; 12:19293-19307. [PMID: 32935695 DOI: 10.1039/d0nr04634e] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Hypoxia in tumor cells is regarded as the most crucial cause of clinical drug resistance and radio-resistance; thus, relieving hypoxia of tumor cells is the key to enhancing the efficacy of anticancer therapy. As a gas signal molecule of vasodilatation factors, nitric oxide (NO) can relieve the hypoxia status of tumor cells, thereby, enhancing the sensitivity of tumor cells to radiotherapy. However, considering complications of vascular activity, the level of NO required for radiotherapy sensitization cannot be obtained in vivo. In view of this, we design and fabricate a multifunctional bismuth-based nanotheranostic agent, which is functionalized with S-nitrosothiol and termed Bi-SNO NPs. X-rays break down the S-N bond and simultaneously trigger large amount of NO-releasing (over 60 μM). Moreover, the as-prepared Bi-SNO NPs not only possess the capability of absorbing and converting 808 nm NIR photons into heat for photothermal therapy, but also have the ability to increase X-ray absorption and CT imaging sensitivity. In addition, the collaborative radio-, photothermal-, and gas-therapy of Bi-SNO in vivo was further investigated and remarkable synergistic tumor inhibition was realized. Finally, no obvious toxicity of Bi-SNO NPs was observed in the treated mice within 14 days. Therefore, the Bi-SNO developed in this work is an effective nano-agent for cancer theranostics with well-controlled morphology and uniform size (36 nm), which could serve as a versatile CT imaging-guided combined radio-, photothermal- and gas-therapy nanocomposite with negligible side effects.
Collapse
Affiliation(s)
- Fangmei Zhang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China.
| | - Shikai Liu
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China.
| | - Na Zhang
- Drug Safety Evaluation Center, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Ye Kuang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China.
| | - Wenting Li
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China.
| | - Shili Gai
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China.
| | - Fei He
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China.
| | - Arif Gulzar
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China.
| | - Piaoping Yang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China.
| |
Collapse
|
14
|
Akakuru OU, Liu C, Iqbal MZ, Dar GI, Yang G, Qian K, Nosike EI, Xing J, Zhang Z, Li Y, Li J, Wu A. A Hybrid Organo-Nanotheranostic Platform of Superlative Biocompatibility for Near-Infrared-Triggered Fluorescence Imaging and Synergistically Enhanced Ablation of Tumors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002445. [PMID: 32954652 DOI: 10.1002/smll.202002445] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/04/2020] [Indexed: 06/11/2023]
Abstract
The quest for an all-organic nanosystem with negligible cytotoxicity and remarkable in vivo tumor theranostic capability is inescapably unending. Hitherto, the landscape of available photothermal agents is dominated by metal-based nanoparticles (NPs) with attendant in vivo negatives. Here, an all-organic-composed theranostic nanosystem with outstanding biocompatibility for fluorescence image-guided tumor photothermal therapy, and as a potential alternative to metal-based photothermal agents is developed. This is rationally achieved by compartmentalizing indocyanine green (ICG) in glycol chitosan (GC)-polypyrrole (PP) nanocarrier to form hybrid ICG@GC-PP NPs (≈65 nm). The compartmentalization strategy, alongside the high photothermal conversion ability of PP jointly enhances the low photostability of free ICG. Advantageously, ICG@GC-PP is endowed with an impeccable in vivo performance by the well-known biocompatibility track records of its individual tri organo-components (GC, PP, and ICG). As a proof of concept, ICG@GC-PP NPs enables a sufficiently prolonged tumor diagnosis by fluorescence imaging up to 20 h post-injection. Furthermore, owing to the complementary heating performances of PP and ICG, ICG@GC-PP NPs-treated mice by one-time near-infrared irradiation exhibit total tumor regression within 14 days post-treatment. Therefore, leveraging the underlying benefits of this study will help to guide the development of new all-organic biocompatible systems in synergism, for safer tumor theranostics.
Collapse
Affiliation(s)
- Ozioma Udochukwu Akakuru
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo, 315201, China
- University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Chuang Liu
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo, 315201, China
- University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - M Zubair Iqbal
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo, 315201, China
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, No. 2 Road of Xiasha, Hangzhou, 310018, China
| | - Gohar Ijaz Dar
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo, 315201, China
- University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Gao Yang
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo, 315201, China
| | - Kun Qian
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo, 315201, China
- University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Elvis Ikechukwu Nosike
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo, 315201, China
- University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Jie Xing
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo, 315201, China
- University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Zhoujing Zhang
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo, 315201, China
| | - Yanying Li
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo, 315201, China
- University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Juan Li
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo, 315201, China
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS) Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, CAS, 1219 ZhongGuan West Road, Ningbo, 315201, China
| |
Collapse
|
15
|
Xiao B, Zhou X, Xu H, Zhang W, Xu X, Tian F, Qian Y, Yu F, Pu C, Hu H, Zhou Z, Liu X, Patra HK, Slater N, Tang J, Gao J, Shen Y. On/off switchable epicatechin-based ultra-sensitive MRI-visible nanotheranostics - see it and treat it. Biomater Sci 2020; 8:5210-5218. [PMID: 32844846 DOI: 10.1039/d0bm00842g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Nanotechnology has a remarkable impact on the preclinical development of future medicines. However, the complicated preparation and systemic toxicity to living systems prevent them from translation to clinical applications. In the present report, we developed a polyepicatechin-based on/off switchable ultra-sensitive magnetic resonance imaging (MRI) visible theranostic nanoparticle (PEMN) for image-guided photothermal therapy (PTT) using our strategy of integrating polymerization and biomineralization into the protein template. We have exploited natural polyphenols as the near infra-red (NIR) switchable photothermal source and MnO2 for the MRI-guided theranostics. PEMN demonstrates excellent MRI contrast ability with a longitudinal relaxivity value up to 30.01 mM-1 s-1. PEMN has shown great tumor inhibition on orthotopic breast tumors and the treatment could be made switchable with an on/off interchangeable mode as needed. PEMN was found to be excretable mainly through the kidneys, avoiding potential systemic toxicity. Thus, PEMN could be extremely useful for developing on-demand therapeutics via'see it and treat it' means with distinguished MRI capability and on/off switchable photothermal properties.
Collapse
Affiliation(s)
- Bing Xiao
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, and College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Wu D, Zhu L, Li Y, Wang H, Xu S, Zhang X, Wu R, Yang G. Superparamagnetic chitosan nanocomplexes for colorectal tumor-targeted delivery of irinotecan. Int J Pharm 2020; 584:119394. [DOI: 10.1016/j.ijpharm.2020.119394] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/11/2020] [Accepted: 04/29/2020] [Indexed: 12/11/2022]
|
17
|
Shan X, Chen Q, Yin X, Jiang C, Li T, Wei S, Zhang X, Sun G, Liu J, Lu L. Polypyrrole-based double rare earth hybrid nanoparticles for multimodal imaging and photothermal therapy. J Mater Chem B 2020; 8:426-437. [DOI: 10.1039/c9tb02254f] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A polypyrrole-based theranostic agent containing double rare-earth elements was constructed and demonstrated promising application for T1/T2-weighted MRI/CT tri-modal imaging guided photothermal cancer therapy.
Collapse
|
18
|
Tao K, Liu S, Wang L, Qiu H, Li B, Zhang M, Guo M, Liu H, Zhang X, Liu Y, Hou Y, Zhang H. Targeted multifunctional nanomaterials with MRI, chemotherapy and photothermal therapy for the diagnosis and treatment of bladder cancer. Biomater Sci 2020; 8:342-352. [PMID: 31724659 DOI: 10.1039/c9bm01377f] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Folate-modified vincristine-loaded polydopamine-coated Fe3O4 superparticles are designed as multifunctional nanomaterials for the imaging and treatment of bladder cancer.
Collapse
|
19
|
Jiang B, Zhang R, Zhang J, Hou Y, Chen X, Zhou M, Tian X, Hao C, Fan K, Yan X. GRP78-targeted ferritin nanocaged ultra-high dose of doxorubicin for hepatocellular carcinoma therapy. Am J Cancer Res 2019; 9:2167-2182. [PMID: 31149036 PMCID: PMC6531302 DOI: 10.7150/thno.30867] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/29/2018] [Indexed: 12/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) remains one of the leading causes of cancer deaths, primarily due to its high incidence of recurrence and metastasis. Considerable efforts have therefore been undertaken to develop effective therapies; however, effective anti-HCC therapies rely on identification of suitable biomarkers, few of which are currently available for drug targeting. Methods: GRP78 was identified as the membrane receptor of HCC-targeted peptide SP94 by immunoprecipitation and mass spectrum analysis. To develop an effective anti-HCC drug nanocarrier, we first displayed GRP78-targeted peptide SP94 onto the exterior surface of Pyrococcus furiosus ferritin Fn (HccFn) by genetic engineering approach, and then loaded doxorubicin (Dox) into the cavities of HccFn via urea-based disassembly/reassembly method, thereby constructing a drug nanocarrier called HccFn-Dox. Results: We demonstrated that HccFn nanocage encapsulated ultra-high dose of Dox (up to 400 molecules Dox/protein nanocage). In vivo animal experiments showed that Dox encapsulated in HccFn-Dox was selectively delivered into HCC tumor cells, and effectively killed subcutaneous and lung metastatic HCC tumors. In addition, HccFn-Dox significantly reduced drug exposure to healthy organs and improved the maximum tolerated dose by six-fold compared with free Dox. Conclusion: In conclusion, our findings clearly demonstrate that GRP78 is an effective biomarker for HCC therapy, and GRP78-targeted HccFn nanocage is effective in delivering anti-HCC drug without damage to healthy tissue.
Collapse
|
20
|
Jin Y, Li Y, Yang X, Tian J. Neuroblastoma-targeting triangular gadolinium oxide nanoplates for precise excision of cancer. Acta Biomater 2019; 87:223-234. [PMID: 30669004 DOI: 10.1016/j.actbio.2019.01.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/03/2019] [Accepted: 01/18/2019] [Indexed: 12/19/2022]
Abstract
Neuroblastoma accounts for 8-10% of malignancies in infants and children. It is urgent to develop an appropriate theranostic agent for effective diagnosis and therapy of neuroblastoma. Herein, we constructed RVG peptide and IRDye800-conjugated bovine serum albumin-coated triangular gadolinium oxide nanoplates (RVG&IRDye800-Gd2O3 TNs) as a targeting MRI agent for the diagnosis of neuroblastoma preoperation and a fluorescence imaging agent for the guidance of the precise excision of the neuroblastoma in surgery. RVG&IRDye800-Gd2O3 TNs have uniform edge length. The RVG&IRDye800-Gd2O3 TNs show remarkably enhanced affinity to both mouse- and human-derived neuroblastoma cells compared with IRDye800-Gd2O3 TNs (3.07-fold and 3.02-fold, respectively). Because of the increased accumulation in tumor cells, RVG&IRDye800-Gd2O3 TNs exhibit signals threefold to fivefold higher than the surrounding normal tissues, which is propitious to the diagnosis of neuroblastoma preoperation and provides real-time visual guidance of the precise excision of the neuroblastoma. Most importantly, with the guidance of the fluorescence imaging agent, the survival rate increased from 0% to 80% 42 days after surgery compared with that in conventional surgery. These findings indicated that the RVG peptide combined with IRDye800-Gd2O3 TNs has the potential to improve the diagnosis and treatment of patients with neuroblastoma. STATEMENT OF SIGNIFICANCE: In this study, we prepared RVG peptide and IRDye800-conjugated bovine serum albumin-coated triangular gadolinium oxide nanoplates (RVG&IRDye800-Gd2O3 TNs) as a targeting MRI agent for the diagnosis of neuroblastoma preoperation and a fluorescence imaging agent for the guidance of the precise excision of the neuroblastoma during surgery. Neuroblastoma was accurately located by MRI imaging, and the tumor margin could be real-time monitored through near-infrared fluorescence imaging. The RVG&IRDye800-Gd2O3 TNs exhibit signals threefold to fivefold higher than those in the surrounding normal tissues, which is propitious to the diagnosis of the neuroblastoma preoperation and provides real-time visual guidance of the precise excision of the neuroblastoma. With the guidance of the fluorescence imaging agent in surgery, the survival rate increased from 0% to 80% 42 days after surgery compared with that in conventional surgery.
Collapse
Affiliation(s)
- Yushen Jin
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Beijing Key Laboratory Molecular Imaging, Beijing 100190, China
| | - Yanyan Li
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Xin Yang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Beijing Key Laboratory Molecular Imaging, Beijing 100190, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Beijing Key Laboratory Molecular Imaging, Beijing 100190, China; The State Key Laboratory of Management and Control for Complex Systems, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100080, China.
| |
Collapse
|
21
|
Jiang B, Yan L, Zhang J, Zhou M, Shi G, Tian X, Fan K, Hao C, Yan X. Biomineralization Synthesis of the Cobalt Nanozyme in SP94-Ferritin Nanocages for Prognostic Diagnosis of Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2019; 11:9747-9755. [PMID: 30777743 DOI: 10.1021/acsami.8b20942] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nanomaterials with intrinsic enzyme-like activities (nanozymes) have emerged as promising agents for cancer theranostics strategies. However, size-controllable synthesis of nanozymes and their targeting modifications are still challenging. Here, we report a monodispersed ferritin-based cobalt nanozyme (HccFn(Co3O4)) that specifically targets and visualizes clinical hepatocellular carcinoma (HCC) tissues. The cobalt nanozyme is biomimetically synthesized within the protein shell of the HCC-targeted ferritin (HccFn) nanocage, which is enabled by the display of HCC cell-specific peptide SP94 on the surface of ferritin through a genetic engineering approach. The intrinsic peroxidase-like activity of HccFn(Co3O4) nanozymes catalyzes the substrates to make color reaction to visualize HCC tumor tissues. We examined 424 clinical HCC specimens and verified that HccFn(Co3O4) nanozymes distinguish HCC tissues from normal liver tissues with a sensitivity of 63.5% and specificity of 79.1%, which is comparable with that of the clinically used HCC-specific marker α fetoprotein. Moreover, the pathological analysis indicates that the HccFn(Co3O4) nanozyme staining result is a potential predictor of prognosis in HCC patients. Staining intensity is positively correlated to tumor differentiation degree ( P = 0.0246) and tumor invasion ( P < 0.0001) and negatively correlated with overall survival ( P = 0.0084) of HCC patients. Together, our study demonstrates that ferritin is an excellent platform for size-controllable synthesis and targeting modifications of nanozymes, and the HccFn(Co3O4) nanozyme is a promising reagent for prognostic diagnosis of HCC.
Collapse
Affiliation(s)
- Bing Jiang
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
- College of Life Sciences , University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Liang Yan
- Key Laboratory of Carcinogenesis and Translational Research, Department of Hepato-Pancreato-Biliary Surgery , Peking University Cancer Hospital & Institute , Beijing 100142 , China
| | - Jianlin Zhang
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
| | - Meng Zhou
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
| | - Guizhi Shi
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
| | - Xiuyun Tian
- Key Laboratory of Carcinogenesis and Translational Research, Department of Hepato-Pancreato-Biliary Surgery , Peking University Cancer Hospital & Institute , Beijing 100142 , China
| | - Kelong Fan
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
| | - Chunyi Hao
- Key Laboratory of Carcinogenesis and Translational Research, Department of Hepato-Pancreato-Biliary Surgery , Peking University Cancer Hospital & Institute , Beijing 100142 , China
| | - Xiyun Yan
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics , Chinese Academy of Sciences , Beijing 100101 , China
- College of Life Sciences , University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
22
|
Doughty ACV, Hoover AR, Layton E, Murray CK, Howard EW, Chen WR. Nanomaterial Applications in Photothermal Therapy for Cancer. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E779. [PMID: 30866416 PMCID: PMC6427777 DOI: 10.3390/ma12050779] [Citation(s) in RCA: 239] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/24/2019] [Accepted: 02/27/2019] [Indexed: 12/13/2022]
Abstract
As a result of their unique compositions and properties, nanomaterials have recently seen a tremendous increase in use for novel cancer therapies. By taking advantage of the optical absorption of near-infrared light, researchers have utilized nanostructures such as carbon nanotubes, gold nanorods, and graphene oxide sheets to enhance photothermal therapies and target the effect on the tumor tissue. However, new uses for nanomaterials in targeted cancer therapy are coming to light, and the efficacy of photothermal therapy has increased dramatically. In this work, we review some of the current applications of nanomaterials to enhance photothermal therapy, specifically as photothermal absorbers, drug delivery vehicles, photoimmunological agents, and theranostic tools.
Collapse
Affiliation(s)
- Austin C V Doughty
- Biophotonics Research Laboratory, Center for Interdisciplinary Biomedical Education and Research, College of Mathematics and Science, University of Central Oklahoma, Edmond, OH 73034, USA.
| | - Ashley R Hoover
- Biophotonics Research Laboratory, Center for Interdisciplinary Biomedical Education and Research, College of Mathematics and Science, University of Central Oklahoma, Edmond, OH 73034, USA.
| | - Elivia Layton
- Biophotonics Research Laboratory, Center for Interdisciplinary Biomedical Education and Research, College of Mathematics and Science, University of Central Oklahoma, Edmond, OH 73034, USA.
| | - Cynthia K Murray
- Department of Mathematics and Statistics, College of Mathematics and Science, University of Central Oklahoma, Edmond, OH 73034, USA.
| | - Eric W Howard
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OH 73104, USA.
| | - Wei R Chen
- Biophotonics Research Laboratory, Center for Interdisciplinary Biomedical Education and Research, College of Mathematics and Science, University of Central Oklahoma, Edmond, OH 73034, USA.
| |
Collapse
|
23
|
Wang Q, Zhang P, Li Z, Feng X, Lv C, Zhang H, Xiao H, Ding J, Chen X. Evaluation of Polymer Nanoformulations in Hepatoma Therapy by Established Rodent Models. Theranostics 2019; 9:1426-1452. [PMID: 30867842 PMCID: PMC6401493 DOI: 10.7150/thno.31683] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/08/2019] [Indexed: 01/10/2023] Open
Abstract
Hepatoma is one of the most severe malignancies usually with poor prognosis, and many patients are insensitive to the existing therapeutic agents, including the drugs for chemotherapy and molecular targeted therapy. Currently, researchers are committed to developing the advanced formulations with controlled drug delivery to improve the efficacy of hepatoma therapy. Numerous inoculated, induced, and genetically engineered hepatoma rodent models are now available for formulation screening. However, animal models of hepatoma cannot accurately represent human hepatoma in terms of histological characteristics, metastatic pathways, and post-treatment responses. Therefore, advanced animal hepatoma models with comparable pathogenesis and pathological features are in urgent need in the further studies. Moreover, the development of nanomedicines has renewed hope for chemotherapy and molecular targeted therapy of advanced hepatoma. As one kind of advanced formulations, the polymer-based nanoformulated drugs have many advantages over the traditional ones, such as improved tumor selectivity and treatment efficacy, and reduced systemic side effects. In this article, the construction of rodent hepatoma model and much information about the current development of polymer nanomedicines were reviewed in order to provide a basis for the development of advanced formulations with clinical therapeutic potential for hepatoma.
Collapse
Affiliation(s)
- Qilong Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Ping Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Zhongmin Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Xiangru Feng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Chengyue Lv
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Huaiyu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| |
Collapse
|
24
|
Fu Q, Zhu R, Song J, Yang H, Chen X. Photoacoustic Imaging: Contrast Agents and Their Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805875. [PMID: 30556205 DOI: 10.1002/adma.201805875] [Citation(s) in RCA: 295] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/10/2018] [Indexed: 05/20/2023]
Abstract
Photoacoustic (PA) imaging as a fast-developing imaging technique has great potential in biomedical and clinical applications. It is a noninvasive imaging modality that depends on the light-absorption coefficient of the imaged tissue and the injected PA-imaging contrast agents. Furthermore, PA imaging provides superb contrast, super spatial resolution, and high penetrability and sensitivity to tissue functional characteristics by detecting the acoustic wave to construct PA images. In recent years, a series of PA-imaging contrast agents are developed to improve the PA-imaging performance in biomedical applications. Here, recent progress of PA contrast agents and their biomedical applications are outlined. PA contrast agents are classified according to their components and function, and gold nanocrystals, gold-nanocrystal assembly, transition-metal chalcogenides/MXene-based nanomaterials, carbon-based nanomaterials, other inorganic imaging agents, small organic molecules, semiconducting polymer nanoparticles, and nonlinear PA-imaging contrast agents are discussed. The applications of PA contrast agents as biosensors (in the sensing of metal ions, pH, enzymes, temperature, hypoxia, reactive oxygen species, and reactive nitrogen species) and in bioimaging (lymph nodes, vasculature, tumors, and brain tissue) are discussed in detail. Finally, an outlook on the future research and investigation of PA-imaging contrast agents and their significance in biomedical research is presented.
Collapse
Affiliation(s)
- Qinrui Fu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Rong Zhu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| |
Collapse
|
25
|
Tumor Photothermal Therapy Employing Photothermal Inorganic Nanoparticles/Polymers Nanocomposites. CHINESE JOURNAL OF POLYMER SCIENCE 2018. [DOI: 10.1007/s10118-019-2193-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
26
|
Li W, Wang X, Wang J, Guo Y, Lu SY, Li CM, Kang Y, Wang ZG, Ran HT, Cao Y, Liu H. Enhanced Photoacoustic and Photothermal Effect of Functionalized Polypyrrole Nanoparticles for Near-Infrared Theranostic Treatment of Tumor. Biomacromolecules 2018; 20:401-411. [DOI: 10.1021/acs.biomac.8b01453] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Wenchao Li
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, Faculty of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Xingyue Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Jingjing Wang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, Faculty of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Yuan Guo
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Shi-Yu Lu
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, Faculty of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Chang Ming Li
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, Faculty of Materials and Energy, Southwest University, Chongqing 400715, China
| | - Yuejun Kang
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, Faculty of Materials and Energy, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Chongqing 400715, China
| | - Zhi-Gang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Hai-Tao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Hui Liu
- Key Laboratory of Luminescent and Real-Time Analytical Chemistry (Southwest University), Ministry of Education, Faculty of Materials and Energy, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Chongqing 400715, China
| |
Collapse
|
27
|
Zhang X, Liu R, Dai Z. Multicolor nanobubbles for FRET/ultrasound dual-modal contrast imaging. NANOSCALE 2018; 10:20347-20353. [PMID: 30375631 DOI: 10.1039/c8nr05488f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The aim of this paper is to develop a novel fluorescence/ultrasound dual-modal contrast agent. We prepared multicolor nanobubbles by doping with three fluorescent dyes for combined fluorescence and contrast enhanced ultrasound imaging. The nanobubbles based on fluorescence resonance energy transfer (FRET) with different doping dye ratio combinations exhibited multiple colors under single wavelength excitation, allowing multiplexed assays for various biomedical applications. In vitro and in vivo ultrasound imaging indicated that nanobubbles have great contrast enhancement capability. In vivo fluorescence imaging showed the excellent ability to provide simultaneous multicolor imaging. The novel multicolor nanobubbles may have great potential for a variety of applications in the study of life science and clinical medicine.
Collapse
Affiliation(s)
- Xiaoting Zhang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China.
| | | | | |
Collapse
|
28
|
Kurbegovic S, Juhl K, Chen H, Qu C, Ding B, Leth JM, Drzewiecki KT, Kjaer A, Cheng Z. Molecular Targeted NIR-II Probe for Image-Guided Brain Tumor Surgery. Bioconjug Chem 2018; 29:3833-3840. [PMID: 30296054 PMCID: PMC6363276 DOI: 10.1021/acs.bioconjchem.8b00669] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Optical
imaging strategies for improving delineation of glioblastoma
(GBM) is highly desired for guiding surgeons to distinguish cancerous
tissue from healthy and precious brain tissue. Fluorescence imaging
(FLI) in the second near-infrared window (NIR-II) outperforms traditional
NIR-I imaging with better tissue penetration, higher spatial and temporal
resolution, and less auto fluorescence and scattering. Because of
high expression in GBM and many other tumors, urokinase Plasminogen
Activator Receptor (uPAR) is an attractive and well proven target
for FLI. Herein we aim to combine the benefit of a NIR-II fluorophore
with a high affinity uPAR targeting small peptide. A targeted NIR-II
fluorescent probe was developed by conjugating an in-house synthesized
NIR-II fluorophore, CH1055, and a uPAR targeting peptide, AE105. To
characterize the in vivo distribution and targeting
properties, a dynamic imaging was performed in orthotopic GBM bearing
nude mice (n = 8). Additionally, fluorescence guided
surgery of orthotopic GBM was performed in living animals. CH1055-4Glu-AE105
was easily synthesized with >75% yield and >98% HPLC evaluated
purity.
The retention time of the probe on analytical HPLC was 15.9 min and
the product was verified by mass spectrometry. Dynamic imaging demonstrated
that the uPAR targeting probe visualized orthotopic GBM through the
intact skull with a tumor-to-background ratio (TBR) of 2.7 peaking
at 96 h. Further, the orthotopic GBM was successfully resected in
small animals guided by the NIR-II FLI. By using a small uPAR targeting
NIR-II probe, FLI allows us to specifically image and detect GBM.
A real-time imaging setup further renders FLI guided tumor resection,
and the probe developed in this work is a promising candidate for
clinical translation.
Collapse
Affiliation(s)
- Sorel Kurbegovic
- Molecular Imaging Program at Stanford (MIPS), Bio-X Program, and Department of Radiology, Canary Center at Stanford for Cancer Early Detection , Stanford University , Stanford , California , 94305-5344 , United States.,Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences , Rigshospitalet and University of Copenhagen , 2200 Copenhagen N, Denmark
| | - Karina Juhl
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences , Rigshospitalet and University of Copenhagen , 2200 Copenhagen N, Denmark
| | - Hao Chen
- Molecular Imaging Program at Stanford (MIPS), Bio-X Program, and Department of Radiology, Canary Center at Stanford for Cancer Early Detection , Stanford University , Stanford , California , 94305-5344 , United States
| | - Chunrong Qu
- Molecular Imaging Program at Stanford (MIPS), Bio-X Program, and Department of Radiology, Canary Center at Stanford for Cancer Early Detection , Stanford University , Stanford , California , 94305-5344 , United States
| | - Bingbing Ding
- Molecular Imaging Program at Stanford (MIPS), Bio-X Program, and Department of Radiology, Canary Center at Stanford for Cancer Early Detection , Stanford University , Stanford , California , 94305-5344 , United States
| | - Julie Maja Leth
- Finsen Laboratory , Rigshospitalet , 2200 Copenhagen N, Denmark.,Biotech Research and Innovation Centre (BRIC) , University of Copenhagen , 2200 Copenhagen N, Denmark
| | - Krzysztof Tadeusz Drzewiecki
- Department of Plastic Surgery, Breast Surgery and Burns Treatment , Rigshospitalet and University of Copenhagen , 2100 Copenhagen Ø, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences , Rigshospitalet and University of Copenhagen , 2200 Copenhagen N, Denmark
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Bio-X Program, and Department of Radiology, Canary Center at Stanford for Cancer Early Detection , Stanford University , Stanford , California , 94305-5344 , United States
| |
Collapse
|