1
|
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
|
2
|
Samaei SS, Daryab M, Gholami S, Rezaee A, Fatehi N, Roshannia R, Hashemi S, Javani N, Rahmanian P, Amani-Beni R, Zandieh MA, Nabavi N, Rashidi M, Malgard N, Hashemi M, Taheriazam A. Multifunctional and stimuli-responsive liposomes in hepatocellular carcinoma diagnosis and therapy. Transl Oncol 2024; 45:101975. [PMID: 38692195 PMCID: PMC11070928 DOI: 10.1016/j.tranon.2024.101975] [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: 12/28/2023] [Revised: 03/11/2024] [Accepted: 04/25/2024] [Indexed: 05/03/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent type of liver cancer, mainly occurring in Asian countries with an increased incidence rate globally. Currently, several kinds of therapies have been deployed for HCC therapy including surgical resection, chemotherapy, radiotherapy and immunotherapy. However, this tumor is still incurable, requiring novel strategies for its treatment. The nanomedicine has provided the new insights regarding the treatment of cancer that liposomes as lipid-based nanoparticles, have been widely applied in cancer therapy due to their biocompaitiblity, high drug loading and ease of synthesis and modification. The current review evaluates the application of liposomes for the HCC therapy. The drugs and genes lack targeting ability into tumor tissues and cells. Therefore, loading drugs or genes on liposomes can increase their accumulation in tumor site for HCC suppression. Moreover, the stimuli-responsive liposomes including pH-, redox- and light-sensitive liposomes are able to deliver drug into tumor microenvironment to improve therapeutic index. Since a number of receptors upregulate on HCC cells, the functionalization of liposomes with lactoferrin and peptides can promote the targeting ability towards HCC cells. Moreover, phototherapy can be induced by liposomes through loading phtoosensitizers to stimulate photothermal- and photodynamic-driven ablation of HCC cells. Overall, the findings are in line with the fact that liposomes are promising nanocarriers for the treatment of HCC.
Collapse
Affiliation(s)
- Seyedeh Setareh Samaei
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mahshid Daryab
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sarah Gholami
- Young Researcher and Elite Club, Babol Branch, Islamic Azad University, Babol, Iran
| | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Navid Fatehi
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Romina Roshannia
- Faculty of Life Science and Bio-technology, Shahid Beheshti University, Tehran, Iran
| | - Saeed Hashemi
- Faculty of Veterinary Medicine, Department of Clinical Sciences, University of Shahrekord, Shahrekord, Iran
| | - Nazanin Javani
- Department of Food Science and Technology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Reza Amani-Beni
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Neda Malgard
- Department of Internal medicine, Firoozgar Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
3
|
Liu T, Liu L, Li L, Cai J. Exploiting targeted nanomedicine for surveillance, diagnosis, and treatment of hepatocellular carcinoma. Mater Today Bio 2023; 22:100766. [PMID: 37636988 PMCID: PMC10457457 DOI: 10.1016/j.mtbio.2023.100766] [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: 05/23/2023] [Revised: 07/26/2023] [Accepted: 08/05/2023] [Indexed: 08/29/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the cancers that has the highest morbidity and mortality rates. In clinical practice, there are still many limitations in surveilling, diagnosing, and treating HCC, such as the poor detection of early HCC, the frequent post-surgery recurrence, the low local tumor control rate, the therapy resistance and side effects. Therefore, improved, or innovative modalities are urgently required for early diagnosis as well as refined and effective management. In recent years, nanotechnology research in the field of HCC has received great attention, with various aspects of diagnosis and treatment including biomarkers, ultrasound, diagnostic imaging, intraoperative imaging, ablation, transarterial chemoembolization, radiotherapy, and systemic therapy. Different from previous reviews that discussed from the perspective of nanoparticles' structure, design and function, this review systematically summarizes the methods and limitations of diagnosing and treating HCC in clinical guidelines and practices, as well as nanomedicine applications. Nanomedicine can overcome the limitations to improve diagnosis accuracy and therapeutic effect via enhancement of targeting, biocompatibility, bioavailability, controlled releasing, and combination of different clinical treatment modalities. Through an in-depth understanding of the logic of nanotechnology to conquer clinical limitations, the main research directions of nanotechnology in HCC are sorted out in this review. It is anticipated that nanomedicine will play a significant role in the future clinical practices of HCC.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Medical Imaging, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510000, China
| | - Li Liu
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, 518000, China
| | - Li Li
- Department of Medical Imaging, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510000, China
| | - Jing Cai
- Department of Medical Imaging, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, 510000, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510000, PR China
| |
Collapse
|
4
|
Feng Y, Chen Q, Jin C, Ruan Y, Chen Q, Lin W, Zhu C, Zhang T, Zhang Y, Gao J, Mo J. Microwave-activated Cu-doped zirconium metal-organic framework for a highly effective combination of microwave dynamic and thermal therapy. J Control Release 2023; 361:102-114. [PMID: 37532150 DOI: 10.1016/j.jconrel.2023.07.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/27/2023] [Accepted: 07/28/2023] [Indexed: 08/04/2023]
Abstract
Percutaneous microwave ablation (PMA) is a thermoablative method used as a minimally invasive treatment for liver cancer. However, the application of PMA is limited by its insufficient ROS generation efficiency and thermal effects. Herein, a new microwave-activated Cu-doped zirconium metal-organic framework (MOF) (CuZr MOF) used for enhanced PMA has a significantly improved microwave sensitizing effect. Owing to the strong inelastic collisions between ions confined in numerous micropores, CuZr MOF has strong microwave sensitivity and high thermal conversion efficiency, which can significantly improve microwave thermal therapy (MTT). Moreover, because of the existence of Cu2+ ions, a further benefit of CuZr MOF is their Fenton-like activity, in particular, microwaves used as an excitation source for microwave dynamic therapy (MDT) can improve the Fenton-like reaction to maximize the synergistic effectiveness of cancer therapy. Importantly, CuZr MOF can inhibit the production of heat shock proteins (HSPs) by producing abundant ROS to enhance tumor destruction. Mechanistically, we found that CuZr MOF + MW treatment modulates ferroptosis-mediated tumor cell death by targeting the HMOX1/GPX4 axis. In summary, this study develops a novel CuZr MOF microwave sensitizer with great potential for synergistic treatment of liver cancer by MTT and MDT.
Collapse
Affiliation(s)
- Yifu Feng
- Department of Hepatobiliary, Taizhou Central Hospital, Taizhou University, Zhejiang 318000, China
| | - Qian Chen
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Chong Jin
- Department of Hepatobiliary, Taizhou Central Hospital, Taizhou University, Zhejiang 318000, China
| | - Yanyun Ruan
- Precision Medicine Center, Taizhou Central Hospital, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Qi Chen
- Precision Medicine Center, Taizhou Central Hospital, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Weidong Lin
- Department of Hepatobiliary, Taizhou Central Hospital, Taizhou University, Zhejiang 318000, China
| | - Chumeng Zhu
- Precision Medicine Center, Taizhou Central Hospital, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200072, China
| | - Yang Zhang
- Nanomedicine and Intestinal Microecology Research Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China; Precision Medicine Center, Taizhou Central Hospital, Taizhou University, Taizhou, Zhejiang 318000, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200072, China.
| | - Jinggang Mo
- Department of Hepatobiliary, Taizhou Central Hospital, Taizhou University, Zhejiang 318000, China.
| |
Collapse
|
5
|
Shi Q, Zhang Z, Zhang W, Ma J, Yang M, Luo J, Liu L, Yan Z. Microwave ablation combined with transarterial chemoembolization containing doxorubicin hydrochloride liposome for treating primary and metastatic liver cancers. J Interv Med 2023; 6:121-125. [PMID: 37846340 PMCID: PMC10577058 DOI: 10.1016/j.jimed.2023.07.002] [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: 04/12/2023] [Revised: 07/05/2023] [Accepted: 07/08/2023] [Indexed: 10/18/2023] Open
Abstract
Aims To determine the safety and efficacy of microwave ablation (MWA) and transarterial chemoembolization (TACE) with doxorubicin hydrochloride liposome (DHL) in patients with primary liver cancer (PLC) and metastatic liver cancer (MLC). Materials and methods The medical records of patients with primary or metastatic liver cancer who underwent MWA combined with TACE containing DHL from March 2019 to March 2022 were collected and analyzed. Treatment-related adverse events (AEs) were recorded. Local tumor response was evaluated according to the modified RECIST criteria. Local tumor progression-free survival (LTPFS) and overall survival (OS) were calculated using the Kaplan-Meier method. Results Altogether, 96 patients with liver cancer were included (PLC, n = 45; MLC, n = 51). Forty (41.7%) patients experienced AEs during treatment, and eight (8.3%) patients developed grade 3 AEs. Compared to before treatment, the serum total bilirubin level and neutrophil to lymphocyte ratio significantly increased after treatment. The median LTPFS was 14.5 months in patients with PLC and 10.7 months in patients with MLC. The median OS was not reached in patients with PLC or MLC. The 1-month and 3-month disease control rates reached more than 80% in both groups. Conclusion MWA combined with TACE with DHL may be a safe and effective method for the treatment of liver cancer.
Collapse
Affiliation(s)
- Qin Shi
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institution of Medical Imaging, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zihan Zhang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institution of Medical Imaging, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wen Zhang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institution of Medical Imaging, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jingqin Ma
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institution of Medical Imaging, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Minjie Yang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institution of Medical Imaging, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jianjun Luo
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institution of Medical Imaging, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lingxiao Liu
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institution of Medical Imaging, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhiping Yan
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institution of Medical Imaging, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
6
|
Wang X, Zhang H, Chen X, Wu C, Ding K, Sun G, Luo Y, Xiang D. Overcoming tumor microenvironment obstacles: Current approaches for boosting nanodrug delivery. Acta Biomater 2023; 166:42-68. [PMID: 37257574 DOI: 10.1016/j.actbio.2023.05.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
In order to achieve targeted delivery of anticancer drugs, efficacy improvement, and side effect reduction, various types of nanoparticles are employed. However, their therapeutic effects are not ideal. This phenomenon is caused by tumor microenvironment abnormalities such as abnormal blood vessels, elevated interstitial fluid pressure, and dense extracellular matrix that affect nanoparticle penetration into the tumor's interstitium. Furthermore, nanoparticle properties including size, charge, and shape affect nanoparticle transport into tumors. This review comprehensively goes over the factors hindering nanoparticle penetration into tumors and describes methods for improving nanoparticle distribution by remodeling the tumor microenvironment and optimizing nanoparticle physicochemical properties. Finally, a critical analysis of future development of nanodrug delivery in oncology is further discussed. STATEMENT OF SIGNIFICANCE: This article reviews the factors that hinder the distribution of nanoparticles in tumors, and describes existing methods and approaches for improving the tumor accumulation from the aspects of remodeling the tumor microenvironment and optimizing the properties of nanoparticles. The description of the existing methods and approaches is followed by highlighting their advantages and disadvantages and put forward possible directions for the future researches. At last, the challenges of improving tumor accumulation in nanomedicines design were also discussed. This review will be of great interest to the broad readers who are committed to delivering nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Xiaohui Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Hong Zhang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Xiaohui Chen
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Chunrong Wu
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Ke Ding
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Guiyin Sun
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China.
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Debing Xiang
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China.
| |
Collapse
|
7
|
Wen L, Liu H, Hu C, Wei Z, Meng Y, Lu C, Su Y, Lu L, Liang H, Xu Q, Zhan M. Thermoacoustic Imaging-Guided Thermo-Chemotherapy for Hepatocellular Carcinoma Sensitized by a Microwave-Responsive Nitric Oxide Nanogenerator. ACS APPLIED MATERIALS & INTERFACES 2023; 15:10477-10491. [PMID: 36790347 DOI: 10.1021/acsami.2c22523] [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: 06/18/2023]
Abstract
Imaging-guided percutaneous microwave thermotherapy has been regarded as an important alternative nonsurgical therapeutic strategy for hepatocellular carcinoma (HCC) that provides excellent local tumor control and favorable survival benefit. However, providing a high-resolution, real-time, and noninvasive imaging technique for intraoperative guidance and controlling postoperative residual tumor recurrence are urgent needs for the clinical setting. In this study, a cisplatin (CDDP)-loaded nanocapsule (NPs@CDDP) with microwave responsive property was prepared to simultaneously serve as a contrast agent of emerging thermoacoustic imaging and a sensitizing agent of microwave thermo-chemotherapy. Accompanying the enzymolysis in the tumor microenvironment, the NPs@CDDP responsively release l-arginine (l-Arg) and CDDP. l-Arg with excellent microwave-absorbing property allowed it to serve as a thermoacoustic imaging contrast agent for accurately delineating the tumor and remarkably increasing tumor temperature under ultralow power microwave irradiation. Apart from the chemotherapeutic effect, CDDP elevated the intracellular H2O2 level through cascade reactions and further accelerated the continuous transformation of l-Arg to nitric oxide (NO), which endowed the NPs@CDDP with NO-generation capability. Notably, the high concentration of intracellular NO was proved to aggravate lipid peroxidation and greatly improved the efficacy of microwave thermo-chemotherapy. Thereby, NPs@CDDP was expected to serve as a theranostic agent integrating the functions of tumor microenvironment-responsive drug delivery system, contrast agent of thermoacoustic imaging, thermal sensitizing agent, and NO nanogenerator, which was promising to provide a potential imaging-guided therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Liewei Wen
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Hongyi Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Cong Hu
- Department of General Surgery, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai 519000, China
| | - Zixuan Wei
- Medical College, Guangxi University, Nanning 530004, China
| | - Ya Meng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR China
| | - Cuixia Lu
- Medical College, Guangxi University, Nanning 530004, China
| | - Yanhong Su
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Hui Liang
- Department of General Surgery, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai 519000, China
| | - Qingbo Xu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
| | - Meixiao Zhan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong 519000, P.R. China
| |
Collapse
|
8
|
Caddeo C, Miglionico R, Rinaldi R, Nigro I, Lamorte D, Chiummiento L, Lupattelli P, Funicello M, D’Orsi R, Valenti D, Santoro V, Fadda AM, Bisaccia F, Vassallo A, Armentano MF. PEGylated Liposomes Loaded with Carbamate Inhibitor ANP0903 Trigger Apoptosis by Enhancing ER Stress in HepG2 Cancer Cells. Int J Mol Sci 2023; 24:ijms24054552. [PMID: 36901980 PMCID: PMC10002784 DOI: 10.3390/ijms24054552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Liver cancer is one of the most common causes of cancer death worldwide. In recent years, substantial progress has been made in the development of systemic therapies, but there is still the need for new drugs and technologies that can increase the survival and quality of life of patients. The present investigation reports the development of a liposomal formulation of a carbamate molecule, reported as ANP0903, previously tested as an inhibitor of HIV-1 protease and now evaluated for its ability to induce cytotoxicity in hepatocellular carcinoma cell lines. PEGylated liposomes were prepared and characterized. Small, oligolamellar vesicles were produced, as demonstrated by light scattering results and TEM images. The physical stability of the vesicles in biological fluids was demonstrated in vitro, alongside the stability during storage. An enhanced cellular uptake was verified in HepG2 cells treated with liposomal ANP0903, resulting in a greater cytotoxicity. Several biological assays were performed to elucidate the molecular mechanisms explaining the proapoptotic effect of ANP0903. Our results allow us to hypothesize that the cytotoxic action in tumor cells is probably due to the inhibition of the proteasome, resulting in an increase in the amount of ubiquitinated proteins within the cells, which in turn triggers activation of autophagy and apoptosis processes, resulting in cell death. The proposed liposomal formulation represents a promising approach to deliver a novel antitumor agent to cancer cells and enhance its activity.
Collapse
Affiliation(s)
- Carla Caddeo
- Department of Scienze della Vita e dell’Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Rocchina Miglionico
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Roberta Rinaldi
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Ilaria Nigro
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy
| | - Lucia Chiummiento
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Paolo Lupattelli
- Department of Chimica, Sapienza University of Roma, p.le Aldo Moro 5, 00185 Roma, Italy
| | - Maria Funicello
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Rosarita D’Orsi
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Donatella Valenti
- Department of Scienze della Vita e dell’Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Valentina Santoro
- Department of Farmacia, University of Salerno, Via Giovanni Paolo II 132, 84084 Salerno, Italy
| | - Anna Maria Fadda
- Department of Scienze della Vita e dell’Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
| | - Faustino Bisaccia
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Antonio Vassallo
- Department of Scienze, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
- Spinoff TNcKILLERS s.r.l., Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
- Correspondence: ; Tel.: +39-0971205624
| | | |
Collapse
|
9
|
Lu Y, Feng N, Du Y, Yu R. Nanoparticle-Based Therapeutics to Overcome Obstacles in the Tumor Microenvironment of Hepatocellular Carcinoma. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12162832. [PMID: 36014696 PMCID: PMC9414814 DOI: 10.3390/nano12162832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 05/09/2023]
Abstract
Hepatocellular carcinoma (HCC) is still a main health concern around the world, with a rising incidence and high mortality rate. The tumor-promoting components of the tumor microenvironment (TME) play a vital role in the development and metastasis of HCC. TME-targeted therapies have recently drawn increasing interest in the treatment of HCC. However, the short medication retention time in TME limits the efficiency of TME modulating strategies. The nanoparticles can be elaborately designed as needed to specifically target the tumor-promoting components in TME. In this regard, the use of nanomedicine to modulate TME components by delivering drugs with protection and prolonged circulation time in a spatiotemporal manner has shown promising potential. In this review, we briefly introduce the obstacles of TME and highlight the updated information on nanoparticles that modulate these obstacles. Furthermore, the present challenges and future prospects of TME modulating nanomedicines will be briefly discussed.
Collapse
Affiliation(s)
- Yuanfei Lu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Na Feng
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
- Correspondence: (Y.D.); (R.Y.); Tel.: +86-571-88208435 (Y.D.); +86-571-87783925 (R.Y.)
| | - Risheng Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, China
- Correspondence: (Y.D.); (R.Y.); Tel.: +86-571-88208435 (Y.D.); +86-571-87783925 (R.Y.)
| |
Collapse
|
10
|
Bartos A, Iancu I, Ciobanu L, Onaciu A, Moldovan C, Moldovan A, Moldovan RC, Tigu AB, Stiufiuc GF, Toma V, Iancu C, Al Hajjar N, Stiufiuc RI. Hybrid Lipid Nanoformulations for Hepatoma Therapy: Sorafenib Loaded Nanoliposomes-A Preliminary Study. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2833. [PMID: 36014698 PMCID: PMC9414144 DOI: 10.3390/nano12162833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 06/15/2023]
Abstract
Sorafenib is a multikinase inhibitor that has received increasing attention due to its high efficacy in hepatocellular carcinoma treatment. However, its poor pharmacokinetic properties (limited water solubility, rapid elimination, and metabolism) still represent major bottlenecks that need to be overcome in order to improve Sorafenib's clinical application. In this paper, we propose a nanotechnology-based hybrid formulation that has the potential to overcome these challenges: sorafenib-loaded nanoliposomes. Sorafenib molecules have been incorporated into the hydrophobic lipidic bilayer during the synthesis process of nanoliposomes using an original procedure developed in our laboratory and, to the best of our knowledge, this is the first paper reporting this type of analysis. The liposomal hybrid formulations have been characterized by transmission electron microscopy (TEM), dynamic light scattering (DLS), and nanoparticle tracking analysis (NTA) that provided useful information concerning their shape, size, zeta-potential, and concentration. The therapeutic efficacy of the nanohybrids has been evaluated on a normal cell line (LX2) and two hepatocarcinoma cell lines, SK-HEP-1 and HepG2, respectively.
Collapse
Affiliation(s)
- Adrian Bartos
- Department of Surgery, Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
- Department of Surgery, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Ioana Iancu
- Department of Surgery, Medicover Hospital, 407062 Cluj-Napoca, Romania
| | - Lidia Ciobanu
- Department of Surgery, Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Anca Onaciu
- MedFuture—Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
- Department of Pharmaceutical Physics-Biophysics, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Cristian Moldovan
- MedFuture—Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
- Department of Pharmaceutical Physics-Biophysics, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Alin Moldovan
- MedFuture—Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Radu Cristian Moldovan
- MedFuture—Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Adrian Bogdan Tigu
- MedFuture—Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | | | - Valentin Toma
- MedFuture—Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Cornel Iancu
- Department of Surgery, Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
| | - Nadim Al Hajjar
- Department of Surgery, Regional Institute of Gastroenterology and Hepatology, 400162 Cluj-Napoca, Romania
- Department of Surgery, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Rares Ionut Stiufiuc
- MedFuture—Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
- Department of Pharmaceutical Physics-Biophysics, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
11
|
Zhao L, Zhang W, Wu Q, Fu C, Ren X, Lv K, Ma T, Chen X, Tan L, Meng X. Lanthanide europium MOF nanocomposite as the theranostic nanoplatform for microwave thermo-chemotherapy and fluorescence imaging. J Nanobiotechnology 2022; 20:133. [PMID: 35292037 PMCID: PMC8922785 DOI: 10.1186/s12951-022-01335-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/26/2022] [Indexed: 12/12/2022] Open
Abstract
Backgrounds Microwave sensitization nanoplatform, integrating multiple functional units for improving tumor selectivity, is of great significance for clinical tumor microwave treatment. Lanthanide europium metal organic framework (EuMOF) is expected to be a theranostic nanoplatform owing to its unique luminescent and microwave sensitization properties. However, it is difficult to be applied to complicated biological systems for EuMOF due to its rapid degradation induced by the solvent molecular and ionic environment. In this work, a luminescent EuMOF nanocomposite (EuMOF@ZIF/AP-PEG, named EZAP) was designed, which brought the multifunctional characteristics of microwave sensitization, fluorescence imaging and drug loading. Results Lamellar EuMOF was synthesized by a hydrothermal method. Through the charge adsorption mechanism, the zeolite imidazole framework (ZIF) structure was intensively assembled on the surface of EuMOF to realize the protection. Then, through in-situ Apatinib drug loading and PEG modification, EZAP nanocomposite was finally obtained. Apatinib (AP) was a kind of chemotherapy drug approved by Food and Drug Administration for targeted therapy of tumors. PEG modification increased long-term circulation of EZAP nanocomposite. The physical and chemical structure and properties of EuMOF@ZIF (EZ) were systematically represented, indicating the successful synthesis of the nanocomposite. The toxic and side effects were negligible at a safe dose. The growth of human liver cancer cells and murine liver cancer cells in vitro was significantly inhibited, and the combined microwave-thermal therapy and chemotherapy in vivo achieved high anti-cancer efficacy. Moreover, EZAP nanocomposite possessed bright red fluorescence, which can be applied for tumor imaging in tumor-bearing mice in vivo. Conclusion Therefore, EZAP nanocomposite showed high microwave sensitization, excellent fluorescence properties and outstanding drug loading capacity, establishing a promising theranostic nanoplatform for tumor therapy and fluorescence imaging. This work proposes a unique strategy to design for the first time a multifunctional nanoplatform with lanthanide metal organic frameworks for biological applications in tumor therapy and diagnosis. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01335-7.
Collapse
Affiliation(s)
- Lirong Zhao
- Laboratory of Controllable Preparation and Application of Nanomaterials, CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29 East Road Zhongguancun, Beijing, 100190, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Wei Zhang
- Department of Interventional Radiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital,, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, People's Republic of China
| | - Qiong Wu
- Laboratory of Controllable Preparation and Application of Nanomaterials, CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29 East Road Zhongguancun, Beijing, 100190, People's Republic of China
| | - Changhui Fu
- Laboratory of Controllable Preparation and Application of Nanomaterials, CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29 East Road Zhongguancun, Beijing, 100190, People's Republic of China
| | - Xiangling Ren
- Laboratory of Controllable Preparation and Application of Nanomaterials, CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29 East Road Zhongguancun, Beijing, 100190, People's Republic of China
| | - Kongpeng Lv
- Department of Interventional Radiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital,, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, People's Republic of China
| | - Tengchuang Ma
- Department of Nuclear Medicine, Harbin Medical University Cancer Hospital, Nangang District, Harbin, 150086, Heilongjiang, People's Republic of China.
| | - Xudong Chen
- Department of Interventional Radiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital,, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, People's Republic of China
| | - Longfei Tan
- Laboratory of Controllable Preparation and Application of Nanomaterials, CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29 East Road Zhongguancun, Beijing, 100190, People's Republic of China.
| | - Xianwei Meng
- Laboratory of Controllable Preparation and Application of Nanomaterials, CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29 East Road Zhongguancun, Beijing, 100190, People's Republic of China.
| |
Collapse
|
12
|
Zhang Y, Guo L, Kong F, Duan L, Li H, Fang C, Zhang K. Nanobiotechnology-enabled energy utilization elevation for augmenting minimally-invasive and noninvasive oncology thermal ablation. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1733. [PMID: 34137183 DOI: 10.1002/wnan.1733] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/15/2021] [Accepted: 05/26/2021] [Indexed: 12/19/2022]
Abstract
Depending on the local or targeted treatment, independence on tumor type and minimally-invasive and noninvasive feature, various thermal ablation technologies have been established, but they still suffer from the intractable paradox between safety and efficacy. It has been extensively accepted that improving energy utilization efficiency is the primary means of decreasing thermal ablation power and shortening ablation time, which is beneficial for concurrently improving both treatment safety and treatment efficiency. Recent efforts have been made to receive a significant advance in various thermal methods including non-invasive high-intensity focused ultrasound, minimally-invasive radiofrequency and microwave, and non-invasive and minimally-invasive photothermal ablation, and so on. Especially, various nanobiotechnologies and design methodologies were employed to elevate the energy utilization efficiency for acquiring unexpected ablation outcomes accompanied with tremendously reduced power and time. More significantly, some combined technologies, for example, chemotherapy, photodynamic therapy (PDT), gaseous therapy, sonodynamic therapy (SDT), immunotherapy, chemodynamic therapy (CDT), or catalytic nanomedicine, were used to assist these ablation means to repress or completely remove tumors. We discussed and summarized the ablation principles and energy transformation pathways of the four ablation means, and reviewed and commented the progress in this field including newly developed technology or new material types with a highlight on nanobiotechnology-inspired design principles, and provided the deep insights into the existing problems and development direction. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Medical Ultrasound, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Lehang Guo
- Department of Medical Ultrasound, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Fanlei Kong
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Lixia Duan
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Hongyan Li
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Chao Fang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China
| | - Kun Zhang
- Department of Medical Ultrasound, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, China.,Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
13
|
Racca L, Cauda V. Remotely Activated Nanoparticles for Anticancer Therapy. NANO-MICRO LETTERS 2020; 13:11. [PMID: 34138198 PMCID: PMC8187688 DOI: 10.1007/s40820-020-00537-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/10/2020] [Indexed: 05/05/2023]
Abstract
Cancer has nowadays become one of the leading causes of death worldwide. Conventional anticancer approaches are associated with different limitations. Therefore, innovative methodologies are being investigated, and several researchers propose the use of remotely activated nanoparticles to trigger cancer cell death. The idea is to conjugate two different components, i.e., an external physical input and nanoparticles. Both are given in a harmless dose that once combined together act synergistically to therapeutically treat the cell or tissue of interest, thus also limiting the negative outcomes for the surrounding tissues. Tuning both the properties of the nanomaterial and the involved triggering stimulus, it is possible furthermore to achieve not only a therapeutic effect, but also a powerful platform for imaging at the same time, obtaining a nano-theranostic application. In the present review, we highlight the role of nanoparticles as therapeutic or theranostic tools, thus excluding the cases where a molecular drug is activated. We thus present many examples where the highly cytotoxic power only derives from the active interaction between different physical inputs and nanoparticles. We perform a special focus on mechanical waves responding nanoparticles, in which remotely activated nanoparticles directly become therapeutic agents without the need of the administration of chemotherapeutics or sonosensitizing drugs.
Collapse
Affiliation(s)
- Luisa Racca
- Department of Applied Science and Technology, Politecnico di Torino, C.so Duca degli Abruzzi 24, 10129, Turin, Italy
| | - Valentina Cauda
- Department of Applied Science and Technology, Politecnico di Torino, C.so Duca degli Abruzzi 24, 10129, Turin, Italy.
| |
Collapse
|