1
|
Fu Y, Zhang Y, Zhang Y, Li R, Yang M, Bai T, Zheng X, Huang D, Zhang M, Tu K, Xu Q, Liu X. Nanoreactors with Cascade Catalytic Activity Reprogram the Tumor Microenvironment for Enhanced Immunotherapy by Synchronously Regulating Treg and Macrophage Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16:49053-49068. [PMID: 39241037 DOI: 10.1021/acsami.4c09830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2024]
Abstract
Immunotherapy has been extensively utilized and studied as a prominent therapeutic strategy for tumors. However, the presence of a hypoxic immunosuppressive tumor microenvironment significantly reduces the efficacy of the treatment, thus impeding its application. In addition, the hypoxic microenvironment can also lead to the enrichment of immunosuppressive cells and reduce the effectiveness of tumor immunotherapy; nanoparticles with biocatalytic activity have the ability to relieve hypoxia in tumor tissues and deliver drugs to target cells and have been widely concerned and applied in the field of tumor therapy. The present study involved the development of a dual nanodelivery system that effectively targets the immune system to modify the tumor microenvironment (TME). The nanodelivery system was developed by incorporating R848 and Imatinib (IMT) into Pt nanozyme loaded hollow polydopamine (P@HP) nanocarriers. Subsequently, their surface was modified with specifically targeted peptides that bind to M2-like macrophages and regulatory T (Treg) cells, thereby facilitating the precise targeting of these cells. When introduced into the tumor model, the nanocarriers were able to selectively target immune cells in tumor tissue, causing M2-type macrophages to change into the M1 phenotype and reducing Treg activation within the tumor microenvironment. In addition, the carriers demonstrated exceptional biocatalytic activity, effectively converting H2O2 into oxygen and water at the tumor site while the drug was active, thereby alleviating the hypoxic inhibitory conditions present in the tumor microenvironment. Additionally, this further enhanced the infiltration of M1-type macrophages and cytotoxic T lymphocytes. Moreover, when used in conjunction with immune checkpoint therapy, the proposed approach demonstrated enhanced antitumor immunotherapeutic effects. The bimodal targeted immunotherapeutic strategy developed in the present study overcomes the drawbacks of traditional immunotherapy approaches while offering novel avenues for the treatment of cancer.
Collapse
Affiliation(s)
- Yuhan Fu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Yuanyuan Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yujie Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Runqing Li
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Mei Yang
- Key Laboratory of Enhanced Recovery after Surgery of Intergrated Chinese and Western Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Ting Bai
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiaoliang Zheng
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang 310053, China
| | - Dongsheng Huang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qiuran Xu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Xin Liu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
2
|
Fei Y, Cao D, Li Y, Wang Z, Dong R, Zhu M, Gao P, Wang X, Cai J, Zuo X. Circ_0008315 promotes tumorigenesis and cisplatin resistance and acts as a nanotherapeutic target in gastric cancer. J Nanobiotechnology 2024; 22:519. [PMID: 39210348 PMCID: PMC11360491 DOI: 10.1186/s12951-024-02760-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Cisplatin-based chemotherapy is one of the fundamental therapeutic modalities for gastric cancer (GC). Chemoresistance to cisplatin is a great clinical challenge, and its underlying mechanisms remain poorly understood. Circular RNAs (circRNAs) are involved in the pathophysiology of multiple human malignancies. METHODS High-throughput sequencing was performed to determine the differentially expressed profile of circRNA in GC tissues and cisplatin-resistant GC cells. Quantitative real-time polymerase chain reaction and Fluorescence in situ hybridization was utilized to confirm the dysregulation of circ_0008315 in GC tissues. To evaluate the prognostic significance of circ_0008315 in GC, we used Kaplan-Meier plot. The self-renewal ability of drug-resistant GC cell was verified through tumor sphere formation assay. GC organoids were constructed to simulate the tumor microenvironment and verified the function of circ_0008315 in cisplatin resistance of gastric cancer. In vivo evaluation was conducted using patient-derived xenograft models. Dual-luciferase reporter gene, RNA immunoprecipitation and miRNA pull-down assays were employed to investigate the molecular mechanisms of circ_0008315 in GC. RESULTS We revealed that a novel circRNA hsa_circ_0008315 was upregulated in GC and cisplatin-resistant GC cells. Elevated circ_0008315 was also observed in cisplatin-resistant GC organoid model. High circ_0008315 expression predicted unfavorable survival outcome in GC patients. Downregulation of circ_0008315 expression inhibited proliferation, mobility, and epithelial-mesenchymal transition of GC cells in vitro and in vivo. Reducing circ_0008315 expression in cisplatin-resistant GC organoid model reversed cisplatin resistance. Mechanistically, circ_0008315 modulated the stem cell properties of GC through the miR-3666/CPEB4 signaling pathway, thereby promoting cisplatin resistance and GC malignant progression. Furthermore, we developed PLGA-PEG nanoparticles targeting circ_0008315, and the nanoparticles could effectively inhibit GC proliferation and cisplatin resistance. CONCLUSION Circ_0008315 exacerbates GC progression and cisplatin resistance, and can be used as a prognostic predictor. Circ_0008315 may function as a promising nanotherapeutic target for GC treatment.
Collapse
Affiliation(s)
- Yao Fei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Danping Cao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Yanna Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Zhixiong Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Runyu Dong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Menglin Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Peng Gao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Xiaoming Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Juan Cai
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China.
- Department of Oncology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China.
| | - Xueliang Zuo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China.
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China.
| |
Collapse
|
3
|
Liu X, Bai Y, Zhou B, Yao W, Song S, Liu J, Zheng C. Recent advances in hepatocellular carcinoma-targeted nanoparticles. Biomed Mater 2024; 19:042004. [PMID: 38697209 DOI: 10.1088/1748-605x/ad46d3] [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: 02/03/2024] [Accepted: 05/01/2024] [Indexed: 05/04/2024]
Abstract
In the field of medicine, we often brave the unknown like interstellar explorers, especially when confronting the formidable opponent of hepatocellular carcinoma (HCC). The global burden of HCC remains significant, with suboptimal treatment outcomes necessitating the urgent development of novel drugs and treatments. While various treatments for liver cancer, such as immunotherapy and targeted therapy, have emerged in recent years, improving their transport and therapeutic efficiency, controlling their targeting and release, and mitigating their adverse effects remains challenging. However, just as we grope through the darkness, a glimmer of light emerges-nanotechnology. Recently, nanotechnology has attracted attention because it can increase the local drug concentration in tumors, reduce systemic toxicity, and has the potential to enhance the effectiveness of precision therapy for HCC. However, there are also some challenges hindering the clinical translation of drug-loaded nanoparticles (NPs). Just as interstellar explorers must overcome interstellar dust, we too must overcome various obstacles. In future researches, the design and development of nanodelivery systems for novel drugs treating HCC should be the first attention. Moreover, researchers should focus on the active targeting design of various NPs. The combination of the interventional therapies and drug-loaded NPs will greatly advance the process of precision HCC therapy.
Collapse
Affiliation(s)
- Xiaoming Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Yaowei Bai
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Binqian Zhou
- Department of Ultrasound, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, People's Republic of China
| | - Wei Yao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Songlin Song
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Jiacheng Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, People's Republic of China
| |
Collapse
|
4
|
Wang Q, Liu J, Chen Z, Zheng J, Wang Y, Dong J. Targeting metabolic reprogramming in hepatocellular carcinoma to overcome therapeutic resistance: A comprehensive review. Biomed Pharmacother 2024; 170:116021. [PMID: 38128187 DOI: 10.1016/j.biopha.2023.116021] [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: 09/18/2023] [Revised: 11/23/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Hepatocellular carcinoma (HCC) poses a heavy burden on human health with high morbidity and mortality rates. Systematic therapy is crucial for advanced and mid-term HCC, but faces a significant challenge from therapeutic resistance, weakening drug effectiveness. Metabolic reprogramming has gained attention as a key contributor to therapeutic resistance. Cells change their metabolism to meet energy demands, adapt to growth needs, or resist environmental pressures. Understanding key enzyme expression patterns and metabolic pathway interactions is vital to comprehend HCC occurrence, development, and treatment resistance. Exploring metabolic enzyme reprogramming and pathways is essential to identify breakthrough points for HCC treatment. Targeting metabolic enzymes with inhibitors is key to addressing these points. Inhibitors, combined with systemic therapeutic drugs, can alleviate resistance, prolong overall survival for advanced HCC, and offer mid-term HCC patients a chance for radical resection. Advances in metabolic research methods, from genomics to metabolomics and cells to organoids, help build the HCC metabolic reprogramming network. Recent progress in biomaterials and nanotechnology impacts drug targeting and effectiveness, providing new solutions for systemic therapeutic drug resistance. This review focuses on metabolic enzyme changes, pathway interactions, enzyme inhibitors, research methods, and drug delivery targeting metabolic reprogramming, offering valuable references for metabolic approaches to HCC treatment.
Collapse
Affiliation(s)
- Qi Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Juan Liu
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 100021, China; Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China; Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing 102218, China; Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, China.
| | - Ziye Chen
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Jingjing Zheng
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yunfang Wang
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 100021, China; Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China; Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing 102218, China; Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China; Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, China.
| | - Jiahong Dong
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Jilin University, Changchun 130021, China; Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing 100021, China; Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China; Institute for Organ Transplant and Bionic Medicine, Tsinghua University, Beijing 102218, China; Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, China.
| |
Collapse
|
5
|
Xia Y, Chen R, Ke Y, Han Q, Ma Z, Shi Q. ROS-responsive phenylboronic ester-based nanovesicles as multifunctional drug delivery systems for the treatment of inflammatory and thrombotic complications. Biomater Sci 2023; 11:7805-7816. [PMID: 37872786 DOI: 10.1039/d3bm01427d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Inflammatory and thrombotic complications and a low loading of dual drugs with different hydrophilicities remain challenges to treat thrombosis with drug delivery systems (DDSs). Here, the reactive oxygen species (ROS)-responsive amphiphilic block polymer poly(ethylene glycol)-b-2-((((4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzyl)oxy)carbonyl)oxy)-ethyl methacrylate (PEG-b-PTBEM) was synthesized and nanovesicles (PPTV) were prepared successfully for the drug delivery platform by controlling the hydrophilic/hydrophobic ratio of molecular chains and molecular self-assembly. The anti-inflammatory drug indomethacin (IDM) was loaded in the wall of nanovesicles and the thrombolytic enzyme nattokinase (NK) was encapsulated in the aqueous cavity of nanovesicles. Both drugs could be rapidly released at the site of thrombosis and/or inflammation with an excessive ROS concentration. The dual drug-loaded nanovesicles not only eliminated ROS, but also alleviated inflammation and dissolved the generated thrombus, showing significant therapeutic efficacy in the in vivo mouse model of carrageenan tail thrombosis. Therefore, drug-delivery nanovesicles play multiple roles in the treatment of inflammation-induced thrombotic disorders, which offer a promising treatment for inflammatory and thrombotic complications.
Collapse
Affiliation(s)
- Yu Xia
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Runhai Chen
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
| | - Yue Ke
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Qiaoyi Han
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Zhifang Ma
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
| | - Qiang Shi
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
- Key Laboratory of Polymeric Materials Design and Synthesis for Biomedical Function, Soochow University, Suzhou, Jiangsu 215123, P. R. China
| |
Collapse
|
6
|
Scotland BL, Shaw JR, Dharmaraj S, Caprio N, Cottingham AL, Joy Martín Lasola J, Sung JJ, Pearson RM. Cell and biomaterial delivery strategies to induce immune tolerance. Adv Drug Deliv Rev 2023; 203:115141. [PMID: 37980950 PMCID: PMC10842132 DOI: 10.1016/j.addr.2023.115141] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023]
Abstract
The prevalence of immune-mediated disorders, including autoimmune conditions and allergies, is steadily increasing. However, current therapeutic approaches are often non-specific and do not address the underlying pathogenic condition, often resulting in impaired immunity and a state of generalized immunosuppression. The emergence of technologies capable of selectively inhibiting aberrant immune activation in a targeted, antigen (Ag)-specific manner by exploiting the body's intrinsic tolerance pathways, all without inducing adverse side effects, holds significant promise to enhance patient outcomes. In this review, we will describe the body's natural mechanisms of central and peripheral tolerance as well as innovative delivery strategies using cells and biomaterials targeting innate and adaptive immune cells to promote Ag-specific immune tolerance. Additionally, we will discuss the challenges and future opportunities that warrant consideration as we navigate the path toward clinical implementation of tolerogenic strategies to treat immune-mediated diseases.
Collapse
Affiliation(s)
- Brianna L Scotland
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Jacob R Shaw
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, United States
| | - Shruti Dharmaraj
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Nicholas Caprio
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Andrea L Cottingham
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Jackline Joy Martín Lasola
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, United States
| | - Junsik J Sung
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, United States; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, United States.
| |
Collapse
|
7
|
Landry J, Shows K, Jagdeesh A, Shah A, Pokhriyal M, Yakovlev V. Regulatory miRNAs in cancer cell recovery from therapy exposure and its implications as a novel therapeutic strategy for preventing disease recurrence. Enzymes 2023; 53:113-196. [PMID: 37748835 DOI: 10.1016/bs.enz.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
The desired outcome of cancer therapies is the eradication of disease. This can be achieved when therapy exposure leads to therapy-induced cancer cell death as the dominant outcome. Theoretically, a permanent therapy-induced growth arrest could also contribute to a complete response, which has the potential to lead to remission. However, preclinical models have shown that therapy-induced growth arrest is not always durable, as recovering cancer cell populations can contribute to the recurrence of cancer. Significant research efforts have been expended to develop strategies focusing on the prevention of recurrence. Recovery of cells from therapy exposure can occur as a result of several cell stress adaptations. These include cytoprotective autophagy, cellular quiescence, a reversable form of senescence, and the suppression of apoptosis and necroptosis. It is well documented that microRNAs regulate the response of cancer cells to anti-cancer therapies, making targeting microRNAs therapeutically a viable strategy to sensitization and the prevention of recovery. We propose that the use of microRNA-targeting therapies in prolonged sequence, that is, a significant period after initial therapy exposure, could reduce toxicity from the standard combination strategy, and could exploit new epigenetic states essential for cancer cells to recover from therapy exposure. In a step toward supporting this strategy, we survey the available scientific literature to identify microRNAs which could be targeted in sequence to eliminate residual cancer cell populations that were arrested as a result of therapy exposure. It is our hope that by successfully identifying microRNAs which could be targeted in sequence we can prevent disease recurrence.
Collapse
Affiliation(s)
- Joseph Landry
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.
| | - Kathryn Shows
- Department of Biology, Virginia State University, Petersburg, VA, United States
| | - Akash Jagdeesh
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Aashka Shah
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Mihir Pokhriyal
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Vasily Yakovlev
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
8
|
Gonsalves A, Sorkhdini P, Bazinet J, Ghumman M, Dhamecha D, Zhou Y, Menon JU. Development and characterization of lung surfactant-coated polymer nanoparticles for pulmonary drug delivery. BIOMATERIALS ADVANCES 2023; 150:213430. [PMID: 37104963 PMCID: PMC10187589 DOI: 10.1016/j.bioadv.2023.213430] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023]
Abstract
Lung cancer is often diagnosed at an advanced stage where tumors are usually inoperable and first-line therapies are inefficient and have off-targeted adverse effects, resulting in poor patient survival. Here, we report the development of an inhalable poly lactic-co-glycolic acid polymer-based nanoparticle (PLGA-NP) formulation with a biomimetic Infasurf® lung surfactant (LS) coating, for localized and sustained lung cancer drug delivery. The nanoparticles (188 ± 7 nm) were stable in phosphate buffered saline, serum and Gamble's solution (simulated lung fluid), and demonstrated cytocompatibility up to 1000 μg/mL concentration and dose-dependent uptake by lung cancer cells. The LS coating significantly decreased nanoparticle (NP) uptake by NR8383 alveolar macrophages in vitro compared to uncoated NPs. The coating, however, did not impair NP uptake by A549 lung adenocarcinoma cells. The anti-cancer drug gemcitabine hydrochloride encapsulated in the PLGA core was released in a sustained manner while the paclitaxel loaded in the LS shell demonstrated a rapid or burst release profile over 21 days. The drug-loaded NPs significantly decreased cancer cell survival and colony formation in vitro compared to free drugs and single drug-loaded NPs. In vivo studies confirmed greater retention of LS-coated NPs in the lungs of C57BL/6 WT mice compared to uncoated NPs, at 24 h and 72 h following intranasal administration. The overall results confirm that LS coating is a unique strategy for cloaking polymeric NPs to potentially prevent their rapid lung clearance and facilitate prolonged pulmonary drug delivery.
Collapse
Affiliation(s)
- Andrea Gonsalves
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Parand Sorkhdini
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Jasmine Bazinet
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Moez Ghumman
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Dinesh Dhamecha
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Yang Zhou
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Jyothi U Menon
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA; Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881, USA.
| |
Collapse
|
9
|
Alqurashi YE, Al-Hetty HRAK, Ramaiah P, Fazaa AH, Jalil AT, Alsaikhan F, Gupta J, Ramírez-Coronel AA, Tayyib NA, Peng H. Harnessing function of EMT in hepatocellular carcinoma: From biological view to nanotechnological standpoint. ENVIRONMENTAL RESEARCH 2023; 227:115683. [PMID: 36933639 DOI: 10.1016/j.envres.2023.115683] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 05/08/2023]
Abstract
Management of cancer metastasis has been associated with remarkable reduction in progression of cancer cells and improving survival rate of patients. Since 90% of mortality are due to cancer metastasis, its suppression can improve ability in cancer fighting. The EMT has been an underlying cause in increasing cancer migration and it is followed by mesenchymal transformation of epithelial cells. HCC is the predominant kind of liver tumor threatening life of many people around the world with poor prognosis. Increasing patient prognosis can be obtained via inhibiting tumor metastasis. HCC metastasis modulation by EMT and HCC therapy by nanoparticles are discussed here. First of all, EMT happens during progression and advanced stages of HCC and therefore, its inhibition can reduce tumor malignancy. Moreover, anti-cancer compounds including all-trans retinoic acid and plumbaging, among others, have been considered as inhibitors of EMT. The EMT association with chemoresistance has been evaluated. Moreover, ZEB1/2, TGF-β, Snail and Twist are EMT modulators in HCC and enhancing cancer invasion. Therefore, EMT mechanism and related molecular mechanisms in HCC are evaluated. The treatment of HCC has not been only emphasized on targeting molecular pathways with pharmacological compounds and since drugs have low bioavailability, their targeted delivery by nanoparticles promotes HCC elimination. Moreover, nanoparticle-mediated phototherapy impairs tumorigenesis in HCC by triggering cell death. Metastasis of HCC and even EMT mechanism can be suppressed by cargo-loaded nanoparticles.
Collapse
Affiliation(s)
- Yaser E Alqurashi
- Department of Biology, College of Science Al-zulfi, Majmaah University, Al-Majmaah, 11952, Saudi Arabia
| | | | | | | | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U. P., India
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Ecuador; Epidemiology and Biostatistics Research Group, CES University, Colombia; Educational Statistics Research Group (GIEE), National University of Education, Ecuador
| | - Nahla A Tayyib
- Faculty of Nursing, Umm Al- Qura University, Makkah, Saudi Arabia
| | - Hu Peng
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
10
|
Sohrab SS, Raj R, Nagar A, Hawthorne S, Paiva-Santos AC, Kamal MA, El-Daly MM, Azhar EI, Sharma A. Chronic Inflammation's Transformation to Cancer: A Nanotherapeutic Paradigm. Molecules 2023; 28:molecules28114413. [PMID: 37298889 DOI: 10.3390/molecules28114413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
The body's normal immune response against any invading pathogen that causes infection in the body results in inflammation. The sudden transformation in inflammation leads to the rise of inflammatory diseases such as chronic inflammatory bowel disease, autoimmune disorders, and colorectal cancer (different types of cancer develop at the site of chronic infection and inflammation). Inflammation results in two ways: short-term inflammation i.e., non-specific, involves the action of various immune cells; the other results in long-term reactions lasting for months or years. It is specific and causes angiogenesis, fibrosis, tissue destruction, and cancer progression at the site of inflammation. Cancer progression relies on the interaction between the host microenvironment and tumor cells along with the inflammatory responses, fibroblast, and vascular cells. The two pathways that have been identified connecting inflammation and cancer are the extrinsic and intrinsic pathways. Both have their own specific role in linking inflammation to cancer, involving various transcription factors such as Nuclear factor kappa B, Activator of transcription, Single transducer, and Hypoxia-inducible factor, which in turn regulates the inflammatory responses via Soluble mediators cytokines (such as Interleukin-6, Hematopoietin-1/Erythropoietin, and tumor necrosis factor), chemokines (such as Cyclooxygenase-2, C-X-C Motif chemokines ligand-8, and IL-8), inflammatory cells, cellular components (such as suppressor cells derived from myeloid, tumor-associated macrophage, and acidophils), and promotes tumorigenesis. The treatment of these chronic inflammatory diseases is challenging and needs early detection and diagnosis. Nanotechnology is a booming field nowadays for its rapid action and easy penetration inside the infected destined cells. Nanoparticles are widely classified into different categories based on their different factors and properties such as size, shape, cytotoxicity, and others. Nanoparticles emerged as excellent with highly progressive medical inventions to cure diseases such as cancer, inflammatory diseases, and others. Nanoparticles have shown higher binding capacity with the biomolecules in inflammation reduction and lowers the oxidative stress inside tissue/cells. In this review, we have overall discussed inflammatory pathways that link inflammation to cancer, major inflammatory diseases, and the potent action of nanoparticles in chronic inflammation-related diseases.
Collapse
Affiliation(s)
- Sayed Sartaj Sohrab
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Riya Raj
- Department of Biochemistry, Bangalore University, Banglore 560056, India
| | - Amka Nagar
- Department of Life Science, School of Basic Science and Research, Sharda University, Greater Noida 201310, India
| | - Susan Hawthorne
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine BT52 1SA, UK
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- LAQV, REQUIMTE, Department of Pharmaceutical Technology, Faculty of Pharmacy of University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Mohammad Amjad Kamal
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Enzymoics Inc., Hebersham, NSW 2770, Australia
- Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
| | - Mai M El-Daly
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Esam I Azhar
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ankur Sharma
- Strathclyde Institute of Pharmaceutical and Biomedical Sciences, University of Strathclyde, Glasgow G1 0RE, UK
| |
Collapse
|
11
|
Qiu C, Wu Y, Shi Q, Guo Q, Zhang J, Meng Y, Wang C, Xia F, Wang J, Xu C. Advanced strategies for nucleic acids and small-molecular drugs in combined anticancer therapy. Int J Biol Sci 2023; 19:789-810. [PMID: 36778126 PMCID: PMC9910002 DOI: 10.7150/ijbs.79328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023] Open
Abstract
Cancer has been considered as complex malignant consequence of genetic mutations that control the cellular proliferation, differentiation and homeostasis, thus making tumor treatment extremely challenging. To date, a variety of cargo molecules, including nucleic acids drugs (pDNA, miRNA and siRNA), therapeutic drugs (doxorubicin, paclitaxel, daunomycin and gefitinib) and imaging agents (radioisotopes, fluorescence dyes, and MRI contrast agents) have been regarded as the potential medicines in clinical application. However, non-single therapeutic drug could induce the satisfied clinical results because of tumor heterogeneity and multiple drug resistance and the nanotechnology-based combined therapy is becoming an advanced important mode for enhanced anticancer effects. The review gathers the current advanced development to co-deliver small-molecular drugs and nucleic acids for the anticancer therapy with nanomedicine-based combination. Furthermore, the superiority is definitely presented and the barriers are detail discussed to surmount the clinical challenges. In final, future perspectives in rational direction for combined tumor therapy of drugs and nucleic acids are exhibited.
Collapse
Affiliation(s)
- Chong Qiu
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yanyan Wu
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Qiaoli Shi
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qiuyan Guo
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Junzhe Zhang
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuqing Meng
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chen Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fei Xia
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jigang Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
- Department of Nephrology, Shenzhen key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, China
| | - Chengchao Xu
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
- College of Integrative Medicine, Laboratory of Pathophysiology, Key Laboratory of Integrative Medicine on Chronic Diseases, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| |
Collapse
|
12
|
MicroRNAs in T Cell-Immunotherapy. Int J Mol Sci 2022; 24:ijms24010250. [PMID: 36613706 PMCID: PMC9820302 DOI: 10.3390/ijms24010250] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/06/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
MicroRNAs (miRNAs) act as master regulators of gene expression in homeostasis and disease. Despite the rapidly growing body of evidence on the theranostic potential of restoring miRNA levels in pre-clinical models, the translation into clinics remains limited. Here, we review the current knowledge of miRNAs as T-cell targeting immunotherapeutic tools, and we offer an overview of the recent advances in miRNA delivery strategies, clinical trials and future perspectives in RNA interference technologies.
Collapse
|
13
|
Zhu S, Li Z, Zheng D, Yu Y, Xiang J, Ma X, Xu D, Qiu J, Yang Z, Wang Z, Li J, Sun H, Chen W, Meng X, Lu Y, Ren Q. A cancer cell membrane coated, doxorubicin and microRNA co-encapsulated nanoplatform for colorectal cancer theranostics. Mol Ther Oncolytics 2022; 28:182-196. [PMID: 36820302 PMCID: PMC9937835 DOI: 10.1016/j.omto.2022.12.002] [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: 02/16/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Endogenous microRNAs (miRNA) in tumors are currently under exhaustive investigation as potential therapeutic agents for cancer treatment. Nevertheless, RNase degradation, inefficient and untargeted delivery, limited biological effect, and currently unclear side effects remain unsettled issues that frustrate clinical application. To address this, a versatile targeted delivery system for multiple therapeutic and diagnostic agents should be adapted for miRNA. In this study, we developed membrane-coated PLGA-b-PEG DC-chol nanoparticles (m-PPDCNPs) co-encapsulating doxorubicin (Dox) and miRNA-190-Cy7. Such a system showed low biotoxicity, high loading efficiency, and superior targeting ability. Systematic delivery of m-PPDCNPs in mouse models showed exceptionally specific tumor accumulation. Sustained release of miR-190 inhibited tumor angiogenesis, tumor growth, and migration by regulating a large group of angiogenic effectors. Moreover, m-PPDCNPs also enhanced the sensitivity of Dox by suppressing TGF-β signal in colorectal cancer cell lines and mouse models. Together, our results demonstrate a stimulating and promising m-PPDCNPs nanoplatform for colorectal cancer theranostics.
Collapse
Affiliation(s)
- Sihao Zhu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Ziyuan Li
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Dongye Zheng
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Yue Yu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jing Xiang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Xiao Ma
- Research Group Signal Transduction, Department of Psychiatry, Ludwig Maximilian University of Munich, Nussbaumstr.7, 80336 Munich, Germany
| | - Dongqing Xu
- Department of Pediatric Hematology/Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jiajun Qiu
- Department of Otolaryngology Head and Neck Surgery, the Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ziyu Yang
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Zhiyi Wang
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Jun Li
- Laboratory Animal Center, Peking University, Beijing 100871, China
| | - Hongfang Sun
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, Gansu Province, China,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou 730000, Gansu Province, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China,NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Beijing 100142, China,Corresponding author.
| | - Yanye Lu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,Institute of Medical Technology, Peking University Health Science Center, Peking University, Beijing 100191, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China,Corresponding author.
| | - Qiushi Ren
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China,National Biomedical Imaging Center, Peking University, Beijing 100871, China,Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen 5181071, China,Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China,Corresponding author.
| |
Collapse
|
14
|
Direct Cardiac Epigenetic Reprogramming through Codelivery of 5'Azacytidine and miR-133a Nanoformulation. Int J Mol Sci 2022; 23:ijms232315179. [PMID: 36499508 PMCID: PMC9739153 DOI: 10.3390/ijms232315179] [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: 07/06/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/09/2022] Open
Abstract
Direct reprogramming of cardiac fibroblasts to induced cardiomyocytes (iCMs) is a promising approach to cardiac regeneration. However, the low yield of reprogrammed cells and the underlying epigenetic barriers limit its potential. Epigenetic control of gene regulation is a primary factor in maintaining cellular identities. For instance, DNA methylation controls cell differentiation in adults, establishing that epigenetic factors are crucial for sustaining altered gene expression patterns with subsequent rounds of cell division. This study attempts to demonstrate that 5'AZA and miR-133a encapsulated in PLGA-PEI nanocarriers induce direct epigenetic reprogramming of cardiac fibroblasts to cardiomyocyte-like cells. The results present a cardiomyocyte-like phenotype following seven days of the co-delivery of 5'AZA and miR-133a nanoformulation into human cardiac fibroblasts. Further evaluation of the global DNA methylation showed a decreased global 5-methylcytosine (5-medCyd) levels in the 5'AZA and 5'AZA/miR-133a treatment group compared to the untreated group and cells with void nanocarriers. These results suggest that the co-delivery of 5'AZA and miR-133a nanoformulation can induce the direct reprogramming of cardiac fibroblasts to cardiomyocyte-like cells in-vitro, in addition to demonstrating the influence of miR-133a and 5'AZA as epigenetic regulators in dictating cell fate.
Collapse
|
15
|
Nano drug delivery systems for antisense oligonucleotides (ASO) therapeutics. J Control Release 2022; 352:861-878. [PMID: 36397636 DOI: 10.1016/j.jconrel.2022.10.050] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 09/02/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022]
Abstract
Cancer, infectious diseases, and metabolic and hereditary genetic disorders are a global health burden affecting millions of people, with contemporary treatments offering limited relief. Antisense technology treats diseases by targeting their causal agents using its ability to alter or inhibit endogenous or malfunctioning genes. Nine antisense oligonucleotide (ASO) drugs that represent four different chemical classes have been approved for the treatment of rare diseases, including nusinersen, the first new oligonucleotide-based drug. Advances in medicinal chemistry, understanding the molecular pathways, and the availability of vast genetic data have resulted in enormous improvements in the therapeutic performance of ASO drugs; however, their susceptibility to degradation in the circulation, rapid renal clearance, and immunostimulatory adverse effects greatly limit their clinical applications. An increasing number of ASO-based therapeutics is being tested in clinical trials. Improvements to the delivery of ASO drugs could potentially change the therapeutic landscape for many conditions in the near future. This review describes the technological advances and developments in drug delivery systems pertaining to ASO therapeutics.
Collapse
|
16
|
Eljack S, David S, Faggad A, Chourpa I, Allard-Vannier E. Nanoparticles design considerations to co-deliver nucleic acids and anti-cancer drugs for chemoresistance reversal. Int J Pharm X 2022; 4:100126. [PMID: 36147518 PMCID: PMC9486027 DOI: 10.1016/j.ijpx.2022.100126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 12/24/2022] Open
Abstract
Chemoresistance and hence the consequent treatment failure is considerably challenging in clinical cancer therapeutics. The understanding of the genetic variations in chemoresistance acquisition encouraged the use of gene modulatory approaches to restore anti-cancer drug efficacy. Many smart nanoparticles are designed and optimized to mediate combinational therapy between nucleic acid and anti-cancer drugs. This review aims to define a rational design of such co-loaded nanocarriers with the aim of chemoresistance reversal at various cellular levels to improve the therapeutic outcome of anticancer treatment. Going through the principles of therapeutics loading, physicochemical characteristics tuning, and different nanocarrier modifications, also looking at combination effectiveness on chemosensitivity restoration. Up to now, these emerging nanocarriers are in development status but are expected to introduce outstanding outcomes.
Collapse
|
17
|
Holjencin C, Jakymiw A. MicroRNAs and Their Big Therapeutic Impacts: Delivery Strategies for Cancer Intervention. Cells 2022; 11:cells11152332. [PMID: 35954176 PMCID: PMC9367537 DOI: 10.3390/cells11152332] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 12/19/2022] Open
Abstract
Three decades have passed from the initial discovery of a microRNA (miRNA) in Caenorhabditis elegans to our current understanding that miRNAs play essential roles in regulating fundamental physiological processes and that their dysregulation can lead to many human pathologies, including cancer. In effect, restoration of miRNA expression or downregulation of aberrantly expressed miRNAs using miRNA mimics or anti-miRNA inhibitors (anti-miRs/antimiRs), respectively, continues to show therapeutic potential for the treatment of cancer. Although the manipulation of miRNA expression presents a promising therapeutic strategy for cancer treatment, it is predominantly reliant on nucleic acid-based molecules for their application, which introduces an array of hurdles, with respect to in vivo delivery. Because naked nucleic acids are quickly degraded and/or removed from the body, they require delivery vectors that can help overcome the many barriers presented upon their administration into the bloodstream. As such, in this review, we discuss the strengths and weaknesses of the current state-of-the-art delivery systems, encompassing viral- and nonviral-based systems, with a specific focus on nonviral nanotechnology-based miRNA delivery platforms, including lipid-, polymer-, inorganic-, and extracellular vesicle-based delivery strategies. Moreover, we also shed light on peptide carriers as an emerging technology that shows great promise in being a highly efficacious delivery platform for miRNA-based cancer therapeutics.
Collapse
Affiliation(s)
- Charles Holjencin
- Department of Oral Health Sciences, James B. Edwards College of Dental Medicine, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA;
| | - Andrew Jakymiw
- Department of Oral Health Sciences, James B. Edwards College of Dental Medicine, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA;
- Department of Biochemistry & Molecular Biology, College of Medicine, Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC 29425, USA
- Correspondence: ; Tel.: +1-843-792-2551
| |
Collapse
|
18
|
Sadeghipour N, Kumar SU, Massoud TF, Paulmurugan R. A rationally identified panel of microRNAs targets multiple oncogenic pathways to enhance chemotherapeutic effects in glioblastoma models. Sci Rep 2022; 12:12017. [PMID: 35835978 PMCID: PMC9283442 DOI: 10.1038/s41598-022-16219-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/06/2022] [Indexed: 11/30/2022] Open
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor. Available treatments have limited success because most patients develop chemoresistance. Alternative strategies are required to improve anticancer effects of current chemotherapeutics while limiting resistance. Successful targeting of microRNAs (miRNAs) as regulators of gene expression can help reprogram GBM cells to better respond to chemotherapy. We aimed to identify a panel of miRNAs that target multiple oncogenic pathways to improve GBM therapy. We first identified differentially expressed miRNAs and tested if their target genes play central roles in GBM signaling pathways by analyzing data in the Gene Expression Omnibus and The Cancer Genome Atlas databases. We then studied the effects of different combinations of these miRNAs in GBM cells by delivering synthetic miRNAs using clinically compatible PLGA-PEG nanoparticles prior to treatment with temozolomide (TMZ) or doxorubicin (DOX). The successful miRNA panel was tested in mice bearing U87-MG cells co-treated with TMZ. We identified a panel of five miRNAs (miRNA-138, miRNA-139, miRNA-218, miRNA-490, and miRNA-21) and their oncogenic targets (CDK6, ZEB1, STAT3, TGIF2, and SMAD7) that cover four different signaling pathways (cell proliferation and apoptotic signaling, invasion and metastasis, cytokine signaling, and stemness) in GBM. We observed significant in vitro and in vivo enhancement of therapeutic efficiency of TMZ and DOX in GBM models. The proposed combination therapy using rationally selected miRNAs and chemotherapeutic drugs is effective owing to the ability of this specific miRNA panel to better target multiple genes associated with the hallmarks of cancer.
Collapse
Affiliation(s)
- Negar Sadeghipour
- Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
- Cellular Pathway Imaging Laboratory (CPIL), The Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sukumar Uday Kumar
- Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA
- Cellular Pathway Imaging Laboratory (CPIL), The Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Tarik F Massoud
- Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA.
- Cellular Pathway Imaging Laboratory (CPIL), The Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA.
- Laboratory of Experimental and Molecular Neuroimaging (LEMNI), Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA, 94304, USA.
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, Stanford, CA, USA.
- Cellular Pathway Imaging Laboratory (CPIL), The Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA.
- Cellular Pathway Imaging Laboratory (CPIL), Molecular Imaging Program at Stanford (MIPS), Canary Center for Cancer Early Detection at Stanford, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA, 94304, USA.
| |
Collapse
|
19
|
Reduced Cardiotoxicity of Ponatinib-Loaded PLGA-PEG-PLGA Nanoparticles in Zebrafish Xenograft Model. MATERIALS 2022; 15:ma15113960. [PMID: 35683259 PMCID: PMC9182153 DOI: 10.3390/ma15113960] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/27/2022] [Accepted: 05/13/2022] [Indexed: 12/04/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) are the new generation of anti-cancer drugs with high potential against cancer cells’ proliferation and growth. However, TKIs are associated with severe cardiotoxicity, limiting their clinical value. One TKI that has been developed recently but not explored much is Ponatinib. The use of nanoparticles (NPs) as a better therapeutic agent to deliver anti-cancer drugs and reduce their cardiotoxicity has been recently considered. In this study, with the aim to reduce Ponatinib cardiotoxicity, Poly(D,L-lactide-co-glycolide)-b-poly(ethyleneoxide)-b-poly(D,L-lactide-co-glycolide) (PLGA-PEG-PLGA) triblock copolymer was used to synthesize Ponatinib in loaded PLGA-PEG-PLGA NPs for chronic myeloid leukemia (CML) treatment. In addition to physicochemical NPs characterization (NPs shape, size, size distribution, surface charge, dissolution rate, drug content, and efficacy of encapsulation) the efficacy and safety of these drug-delivery systems were assessed in vivo using zebrafish. Zebrafish are a powerful animal model for investigating the cardiotoxicity associated with anti-cancer drugs such as TKIs, to determine the optimum concentration of smart NPs with the least side effects, and to generate a xenograft model of several cancer types. Therefore, the cardiotoxicity of unloaded and drug-loaded PLGA-PEG-PLGA NPs was studied using the zebrafish model by measuring the survival rate and cardiac function parameters, and therapeutic concentration for in vivo efficacy studies was optimized in an in vivo setting. Further, the efficacy of drug-loaded PLGA-PEG-PLGA NPs was tested on the zebrafish cancer xenograft model, in which human myelogenous leukemia cell line K562 was transplanted into zebrafish embryos. Our results demonstrated that the Ponatinib-loaded PLGA-PEG-PLGA NPs at a concentration of 0.001 mg/mL are non-toxic/non-cardio-toxic in the studied zebrafish xenograft model.
Collapse
|
20
|
Bhattacharjee S. Craft of Co-encapsulation in Nanomedicine: A Struggle To Achieve Synergy through Reciprocity. ACS Pharmacol Transl Sci 2022; 5:278-298. [PMID: 35592431 PMCID: PMC9112416 DOI: 10.1021/acsptsci.2c00033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Indexed: 12/19/2022]
Abstract
Achieving synergism, often by combination therapy via codelivery of chemotherapeutic agents, remains the mainstay of treating multidrug-resistance cases in cancer and microbial strains. With a typical core-shell architecture and surface functionalization to ensure facilitated targeting of tissues, nanocarriers are emerging as a promising platform toward gaining such synergism. Co-encapsulation of disparate theranostic agents in nanocarriers-from chemotherapeutic molecules to imaging or photothermal modalities-can not only address the issue of protecting the labile drug payload from a hostile biochemical environment but may also ensure optimized drug release as a mainstay of synergistic effect. However, the fate of co-encapsulated molecules, influenced by temporospatial proximity, remains unpredictable and marred with events with deleterious impact on therapeutic efficacy, including molecular rearrangement, aggregation, and denaturation. Thus, more than just an art of confining multiple therapeutics into a 3D nanoscale space, a co-encapsulated nanocarrier, while aiming for synergism, should strive toward achieving a harmonious cohabitation of the encapsulated molecules that, despite proximity and opportunities for interaction, remain innocuous toward each other and ensure molecular integrity. This account will inspect the current progress in co-encapsulation in nanocarriers and distill out the key points toward accomplishing such synergism through reciprocity.
Collapse
Affiliation(s)
- Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
21
|
Mitochondrial-Related Transcriptome Feature Correlates with Prognosis, Vascular Invasion, Tumor Microenvironment, and Treatment Response in Hepatocellular Carcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1592905. [PMID: 35535359 PMCID: PMC9078845 DOI: 10.1155/2022/1592905] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/30/2022] [Indexed: 01/17/2023]
Abstract
Background Hepatocellular carcinoma (HCC) is the most common subtype of primary liver cancer, which was highly correlated with metabolic dysfunction. Nevertheless, the association between nuclear mitochondrial-related transcriptome and HCC remained unclear. Materials and Methods A total of 147 nuclear mitochondrial-related genes (NMRGs) were downloaded from the MITOMAP: A Human Mitochondrial Genome Database. The training dataset was downloaded from The Cancer Genome Atlas (TCGA), while validation datasets were retrieved from the International Cancer Genome Consortium (ICGC) and Gene Expression Omnibus (GEO). The univariate and multivariate, and least absolute shrinkage and selection operator (LASSO) Cox regression analyses were applied to construct a NMRG signature, and the value of area under receiver operating characteristic curve (AUC) was utilized to assess the signature and nomogram. Then, data from the Genomics of Drug Sensitivity in Cancer (GDSC) were used for the evaluation of chemotherapy response in HCC. Results Functional enrichment of differentially expressed genes (DEGs) between HCC and paired normal tissue samples demonstrated that mitochondrial dysfunction was significantly associated with HCC development. Survival analysis showed a total of 35 NMRGs were significantly correlated with overall survival (OS) of HCC, and the LASSO Cox regression analysis further identified a 25-NMRG signature and corresponding prognosis score based on their transcriptional profiling. HCC patients were divided into high- and low-risk groups according to the median prognosis score, and high-risk patients had significantly worse OS (median OS: 27.50 vs. 83.18 months, P < 0.0001). The AUC values for OS at 1, 3, and 5 years were 0.79, 0.77, and 0.77, respectively. The prognostic capacity of NMRG signature was verified in the GSE14520 dataset and ICGC-HCC cohort. Besides, the NMRG signature outperformed each NMRG and clinical features in prognosis prediction and could also differentiate whether patients presented with vascular invasions (VIs) or not. Subsequently, a prognostic nomogram (C-index: 0.753, 95% CI: 0.703~0.804) by the integration of age, tumor metastasis, and NMRG prognosis score was constructed with the AUC values for OS at 1, 3, and 5 years were 0.82, 0.81, and 0.82, respectively. Notably, significant enrichment of regulatory and follicular helper T cells in high-risk group indicated the potential treatment of immune checkpoint inhibitors for these patients. Interestingly, the NMRG signature could also identify the potential responders of sorafenib or transcatheter arterial chemoembolization (TACE) treatment. Additionally, HCC patients in high-risk group appeared to be more sensitive to cisplatin, vorinostat, and methotrexate, reversely, patients in low-risk group had significantly higher sensitivity to paclitaxel and bleomycin instead. Conclusions In summary, the development of NMRG signature provided a more comprehensive understanding of mitochondrial dysfunction in HCC, helped predict prognosis and tumor microenvironment, and provided potential targeted therapies for HCC patients with different NMRG prognosis scores.
Collapse
|
22
|
Khare S, Khare T, Ramanathan R, Ibdah JA. Hepatocellular Carcinoma: The Role of MicroRNAs. Biomolecules 2022; 12:biom12050645. [PMID: 35625573 PMCID: PMC9138333 DOI: 10.3390/biom12050645] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths worldwide. HCC is diagnosed in its advanced stage when limited treatment options are available. Substantial morphologic, genetic and epigenetic heterogeneity has been reported in HCC, which poses a challenge for the development of a targeted therapy. In this review, we discuss the role and involvement of several microRNAs (miRs) in the heterogeneity and metastasis of hepatocellular carcinoma with a special emphasis on their possible role as a diagnostic and prognostic tool in the risk prediction, early detection, and treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Sharad Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65212, USA; (S.K.); (T.K.); (R.R.)
- Harry S. Truman Veterans Hospital, Columbia, MO 65201, USA
| | - Tripti Khare
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65212, USA; (S.K.); (T.K.); (R.R.)
| | - Raghu Ramanathan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65212, USA; (S.K.); (T.K.); (R.R.)
- Harry S. Truman Veterans Hospital, Columbia, MO 65201, USA
| | - Jamal A. Ibdah
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO 65212, USA; (S.K.); (T.K.); (R.R.)
- Harry S. Truman Veterans Hospital, Columbia, MO 65201, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
- Correspondence: ; Tel.: 1-573-882-7349; Fax: 1-573-884-4595
| |
Collapse
|
23
|
Li H, Yang YG, Sun T. Nanoparticle-Based Drug Delivery Systems for Induction of Tolerance and Treatment of Autoimmune Diseases. Front Bioeng Biotechnol 2022; 10:889291. [PMID: 35464732 PMCID: PMC9019755 DOI: 10.3389/fbioe.2022.889291] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 11/13/2022] Open
Abstract
Autoimmune disease is a chronic inflammatory disease caused by disorders of immune regulation. Antigen-specific immunotherapy has the potential to inhibit the autoreactivity of inflammatory T cells and induce antigen-specific immune suppression without impairing normal immune function, offering an ideal strategy for autoimmune disease treatment. Tolerogenic dendritic cells (Tol DCs) with immunoregulatory functions play important roles in inducing immune tolerance. However, the effective generation of tolerogenic DCs in vivo remains a great challenge. The application of nanoparticle-based drug delivery systems in autoimmune disease treatment can increase the efficiency of inducing antigen-specific tolerance in vivo. In this review, we discuss multiple nanoparticles, with a focus on their potential in treatment of autoimmune diseases. We also discuss how the physical properties of nanoparticles influence their therapeutic efficacy.
Collapse
Affiliation(s)
- He Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- Department of Rehabilitation Medicine, The First Hospital, Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| |
Collapse
|
24
|
Yang S, Cai C, Wang H, Ma X, Shao A, Sheng J, Yu C. Drug delivery strategy in hepatocellular carcinoma therapy. Cell Commun Signal 2022; 20:26. [PMID: 35248060 PMCID: PMC8898478 DOI: 10.1186/s12964-021-00796-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 10/20/2021] [Indexed: 12/15/2022] Open
Abstract
AbstractHepatocellular carcinoma (HCC) is one of the most common malignant tumors worldwide, with high rates of recurrence and death. Surgical resection and ablation therapy have limited efficacy for patients with advanced HCC and poor liver function, so pharmacotherapy is the first-line option for those patients. Traditional antitumor drugs have the disadvantages of poor biological distribution and pharmacokinetics, poor target selectivity, high resistance, and high toxicity to nontargeted tissues. Recently, the development of nanotechnology has significantly improved drug delivery to tumor sites by changing the physical and biological characteristics of drugs and nanocarriers to improve their pharmacokinetics and biological distribution and to selectively accumulate cytotoxic agents at tumor sites. Here, we systematically review the tumor microenvironment of HCC and the recent application of nanotechnology in HCC.
Collapse
|
25
|
Sayyed AA, Gondaliya P, Bhat P, Mali M, Arya N, Khairnar A, Kalia K. Role of miRNAs In Cancer Diagnostics And Therapy: A Recent Update. Curr Pharm Des 2021; 28:471-487. [PMID: 34751112 DOI: 10.2174/1381612827666211109113305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 07/16/2021] [Indexed: 12/15/2022]
Abstract
The discovery of miRNAs has been one of the revolutionary developments and has led to the advent of new diagnostic and therapeutic opportunities for the management of cancer. In this regard, miRNA dysregulation has been shown to play a critical role in various stages of tumorigenesis, including tumor invasion, metastasis as well as angiogenesis. Therefore, miRNA profiling can provide accurate fingerprints for the development of diagnostic and therapeutic platforms. This review discusses the recent discoveries of miRNA-based tools for early detection of cancer as well as disease monitoring in cancers that are common, like breast, lung, hepatic, colorectal, oral and brain cancer. Based on the involvement of miRNA in different cancers as oncogenic miRNA or tumor suppressor miRNA, the treatment with miRNA inhibitors or mimics is recommended. However, the stability and targeted delivery of miRNA remain the major limitations of miRNA delivery. In relation to this, several nanoparticle-based delivery systems have been reported which have effectively delivered the miRNA mimics or inhibitors and showed the potential for transforming these advanced delivery systems from bench to bedside in the treatment of cancer metastasis and chemoresistance. Based on this, we attempted to uncover recently reported advanced nanotherapeutic approaches to deliver the miRNAs in the management of different cancers.
Collapse
Affiliation(s)
- Adil A Sayyed
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat. India
| | - Piyush Gondaliya
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat. India
| | - Palak Bhat
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat. India
| | - Mukund Mali
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat. India
| | - Neha Arya
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat. India
| | - Amit Khairnar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat. India
| | - Kiran Kalia
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat. India
| |
Collapse
|
26
|
Fawzi Kabil M, Nasr M, El-Sherbiny IM. Conventional and hybrid nanoparticulate systems for the treatment of hepatocellular carcinoma: An updated review. Eur J Pharm Biopharm 2021; 167:9-37. [PMID: 34271117 DOI: 10.1016/j.ejpb.2021.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/28/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is considered a serious malignancy which affects a large number of people worldwide. Despite the presence of some diagnostic techniques for HCC, the fact that its symptoms somehow overlap with other diseases causes it to be diagnosed at a late stage, hence negatively affecting the prognosis of the disease. The currently available treatment strategies have many shortcomings such as high cost, induction of serious side effects as well as multiple drug resistance, hence resulting in therapeutic failure. Accordingly, nanoformulations have been developed in order to overcome the clinical challenges, enhance the therapeutic efficacy, and elicit chemotherapy tailor-ability. Hybrid nanoparticulate carriers in particular, which are composed of two or more drug vehicles with different physicochemical characteristics combined together in one system, have been recently reported to advance nanotechnology-based therapies. Therefore, this review sheds the light on HCC, and the role of nanotechnology and hybrid nanoparticulate carriers as well as the latest developments in the use of conventional nanoparticles in combating this disease.
Collapse
Affiliation(s)
- Mohamed Fawzi Kabil
- Center for Materials Science, University of Science and Technology, Zewail City of Science and Technology, 6th October City, Giza 12578, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ibrahim M El-Sherbiny
- Center for Materials Science, University of Science and Technology, Zewail City of Science and Technology, 6th October City, Giza 12578, Egypt.
| |
Collapse
|
27
|
Therapeutic strategies for miRNA delivery to reduce hepatocellular carcinoma. Semin Cell Dev Biol 2021; 124:134-144. [PMID: 33926792 DOI: 10.1016/j.semcdb.2021.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/21/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022]
Abstract
Malignancies of hepatocellular carcinoma (HCC) are rapidly spreading and commonly fatal. Like most cancers, the gene expression patterns in HCC vary significantly from patient to patient. Moreover, the expression networks during HCC progression are largely controlled by microRNAs (miRNAs) regulating multiple oncogenes and tumor supressors. Therefore, miRNA-based therapeutic strategies altering these networks may significantly influence the cellular behavior enough for them to cure HCC. However, the most substantial challenges in developing such therapies are the stability of the oligos themselves and that of their delivery systems. Here we provide a comprehensive update describing various miRNA delivery systems, including virus-based delivery and non-viral delivery. The latter may be achieved using inorganic nanoparticles, polymer based nano-carriers, lipid-based vesicles, exosomes, and liposomes. Leaky vasculature in HCC-afflicted livers helps untargeted nanocarriers to accumulate in the tumor tissue but may result in side effects during higher dose of treatment. On the other hand, the strategies for actively targeting miRNA therepeutics to cancerous cells through nano-conjugates or vesicles by decorating their surface with antibodies against or ligands for HCC-specific antigens or receptors are more efficient in preventing damage to healthy tissue and cancer recurrence.
Collapse
|
28
|
Grixti JM, Ayers D, Day PJR. An Analysis of Mechanisms for Cellular Uptake of miRNAs to Enhance Drug Delivery and Efficacy in Cancer Chemoresistance. Noncoding RNA 2021; 7:27. [PMID: 33923485 PMCID: PMC8167612 DOI: 10.3390/ncrna7020027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
Up until recently, it was believed that pharmaceutical drugs and their metabolites enter into the cell to gain access to their targets via simple diffusion across the hydrophobic lipid cellular membrane, at a rate which is based on their lipophilicity. An increasing amount of evidence indicates that the phospholipid bilayer-mediated drug diffusion is in fact negligible, and that drugs pass through cell membranes via proteinaceous membrane transporters or carriers which are normally used for the transportation of nutrients and intermediate metabolites. Drugs can be targeted to specific cells and tissues which express the relevant transporters, leading to the design of safe and efficacious treatments. Furthermore, transporter expression levels can be manipulated, systematically and in a high-throughput manner, allowing for considerable progress in determining which transporters are used by specific drugs. The ever-expanding field of miRNA therapeutics is not without its challenges, with the most notable one being the safe and effective delivery of the miRNA mimic/antagonist safely to the target cell cytoplasm for attaining the desired clinical outcome, particularly in miRNA-based cancer therapeutics, due to the poor efficiency of neo-vascular systems revolting around the tumour site, brought about by tumour-induced angiogenesis. This acquisition of resistance to several types of anticancer drugs can be as a result of an upregulation of efflux transporters expression, which eject drugs from cells, hence lowering drug efficacy, resulting in multidrug resistance. In this article, the latest available data on human microRNAs has been reviewed, together with the most recently described mechanisms for miRNA uptake in cells, for future therapeutic enhancements against cancer chemoresistance.
Collapse
Affiliation(s)
- Justine M. Grixti
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Biosciences Building, University of Liverpool, Liverpool L69 7ZB, UK;
| | - Duncan Ayers
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD 2080, Malta
- Faculty of Biology, Medicine and Human Sciences, The University of Manchester, Manchester M1 7DN, UK;
| | - Philip J. R. Day
- Faculty of Biology, Medicine and Human Sciences, The University of Manchester, Manchester M1 7DN, UK;
| |
Collapse
|
29
|
Song C, Xiao Y, Ouyang Z, Shen M, Shi X. Efficient co-delivery of microRNA 21 inhibitor and doxorubicin to cancer cells using core-shell tecto dendrimers formed via supramolecular host-guest assembly. J Mater Chem B 2021; 8:2768-2774. [PMID: 32154812 DOI: 10.1039/d0tb00346h] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Development of versatile and powerful nanoplatforms for efficient therapeutic delivery represents a major topic for current nanomedicine. Herein, we present the development of core-shell tecto dendrimers (CSTDs) for co-delivery of a therapeutic gene and drug for enhanced anticancer therapy applications. In this work, CSTDs were first prepared via supramolecular recognition of β-cyclodextrin (CD)-decorated generation 5 (G5) poly(amidoamine) (PAMAM) dendrimers as cores and adamantane (Ad)-functionalized G3 PAMAM dendrimers as shell components. The formed CSTDs with each G5 dendrimer surrounded with 4.2 G3 dendrimers were evaluated as a gene vector for delivery of plasmid DNA encoding enhanced green fluorescent protein as well as microRNA 21 inhibitor (miR 21i). We show that under an appropriate N/P ratio, the CSTDs enable effective transfection of both genetic materials to cancer cells. In particular, the transfection of miR 21i led to the inhibition of cancer cell migration, decreased miR 21 gene expression, and the effective regulation of the target genes and proteins (e.g., PTEN, PDCD4, p53, and Caspase-3). Furthermore, we revealed that the CSTDs were able to co-deliver miR 21i and an anticancer drug doxorubicin, leading to enhanced therapeutic efficacy to cancer cells in vitro. Our findings imply that the developed CSTDs could be adopted as a versatile platform for effective co-delivery of different therapeutic components for enhanced anticancer therapy applications.
Collapse
Affiliation(s)
- Cong Song
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-dimension Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P. R. China.
| | - Yunchao Xiao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-dimension Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P. R. China.
| | - Zhijun Ouyang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-dimension Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P. R. China.
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-dimension Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P. R. China.
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, International Joint Laboratory for Advanced Fiber and Low-dimension Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P. R. China. and CQM-Centro de Quimica da Madeira, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal
| |
Collapse
|
30
|
Damase TR, Sukhovershin R, Boada C, Taraballi F, Pettigrew RI, Cooke JP. The Limitless Future of RNA Therapeutics. Front Bioeng Biotechnol 2021; 9:628137. [PMID: 33816449 PMCID: PMC8012680 DOI: 10.3389/fbioe.2021.628137] [Citation(s) in RCA: 297] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/15/2021] [Indexed: 12/19/2022] Open
Abstract
Recent advances in the generation, purification and cellular delivery of RNA have enabled development of RNA-based therapeutics for a broad array of applications. RNA therapeutics comprise a rapidly expanding category of drugs that will change the standard of care for many diseases and actualize personalized medicine. These drugs are cost effective, relatively simple to manufacture, and can target previously undruggable pathways. It is a disruptive therapeutic technology, as small biotech startups, as well as academic groups, can rapidly develop new and personalized RNA constructs. In this review we discuss general concepts of different classes of RNA-based therapeutics, including antisense oligonucleotides, aptamers, small interfering RNAs, microRNAs, and messenger RNA. Furthermore, we provide an overview of the RNA-based therapies that are currently being evaluated in clinical trials or have already received regulatory approval. The challenges and advantages associated with use of RNA-based drugs are also discussed along with various approaches for RNA delivery. In addition, we introduce a new concept of hospital-based RNA therapeutics and share our experience with establishing such a platform at Houston Methodist Hospital.
Collapse
Affiliation(s)
- Tulsi Ram Damase
- RNA Therapeutics Program, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Roman Sukhovershin
- RNA Therapeutics Program, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Christian Boada
- Colleges of Medicine, Engineering, Texas A&M University and Houston Methodist Hospital, Houston, TX, United States
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX, United States
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Roderic I. Pettigrew
- Colleges of Medicine, Engineering, Texas A&M University and Houston Methodist Hospital, Houston, TX, United States
| | - John P. Cooke
- RNA Therapeutics Program, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
31
|
Wang Q, Xiao J, Su Y, Huang J, Li J, Qiu L, Zhan M, He X, Yuan W, Li Y. Fabrication of thermoresponsive magnetic micelles from amphiphilic poly(phenyl isocyanide) and Fe3O4 nanoparticles for controlled drug release and synergistic thermochemotherapy. Polym Chem 2021. [DOI: 10.1039/d1py00022e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The drug-loaded micelles self-assembled from co-poly(phenyl isocyanide), Fe3O4 and DOX demonstrated thermoresponsiveness and magnetic hyperthermia for synergistic thermochemotherapy.
Collapse
|
32
|
Paroha S, Verma J, Dubey RD, Dewangan RP, Molugulu N, Bapat RA, Sahoo PK, Kesharwani P. Recent advances and prospects in gemcitabine drug delivery systems. Int J Pharm 2021; 592:120043. [DOI: 10.1016/j.ijpharm.2020.120043] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/17/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022]
|
33
|
Kumar V, Rahman M, Gahtori P, Al-Abbasi F, Anwar F, Kim HS. Current status and future directions of hepatocellular carcinoma-targeted nanoparticles and nanomedicine. Expert Opin Drug Deliv 2020; 18:673-694. [PMID: 33295218 DOI: 10.1080/17425247.2021.1860939] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a major health problem worldwide. Conventional therapies covering either chemotherapy or combination therapy still have sub-optimal responses with significant adverse effects and toxicity. Moreover, tumor cells usually acquire resistance quickly for traditional approaches, limiting their use in HCC. Interest in nanomedicine due to minimal systemic toxicity and a high degree of target-specific drug-delivery have pulled the attention of health scientists in this area of therapeutics. AREA COVERED The review covers the incidence and epidemiology of HCC, proposed molecular drug targets, mechanistic approach and emergence of nanomedicines including nanoparticles, lipidic nanoparticles, vesicular-based nanocarrier, virus-like particles with momentous therapeutic aspects including biocompatibility, and toxicity of nanocarriers along with conclusions and future perspective, with an efficient approach to safely cross physiological barriers to reach the target site for treating liver cancer. EXPERT OPINION Remarkable outcomes have recently been observed for the therapeutic efficacy of nanocarriers with respect to a specific drug target against the treatment of HCC by existing under trial drugs.
Collapse
Affiliation(s)
- Vikas Kumar
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Sciences, Shalom Institute of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, India
| | - Mahfoozur Rahman
- Natural Product Drug Discovery Laboratory, Department of Pharmaceutical Sciences, Shalom Institute of Health Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, India
| | - Prashant Gahtori
- School of Pharmacy, Graphic Era Hill University Dehradun 248002, Uttarakhand, India
| | - Fahad Al-Abbasi
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Firoz Anwar
- Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Jangan-gu, Suwon 16419, 2066, Seobu-ro, Korea
| |
Collapse
|
34
|
Jagwani S, Jalalpure S, Dhamecha D, Jadhav K, Bohara R. Pharmacokinetic and Pharmacodynamic Evaluation of Resveratrol Loaded Cationic Liposomes for Targeting Hepatocellular Carcinoma. ACS Biomater Sci Eng 2020; 6:4969-4984. [PMID: 33455290 DOI: 10.1021/acsbiomaterials.0c00429] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide. The destructive nature of the disease makes it difficult for clinicians to manage the condition. Hence, there is an urgent need to find new alternatives for HCC, as the role of conventional cytotoxic drugs has reached a plateau to control HCC associated mortality. Antioxidant compounds of plant origin with potential anti-tumor effect have been recognized as alternate modes in cancer treatment and chemoprevention. Resveratrol (RS) is a model natural nonflavonoid drug known for its anti-cancer activity. However, its clinical application is limited due to its poor bioavailability. The current research work aims to formulate, optimize, and characterize RS loaded cationic liposomes (RLs) for specific delivery in HCC. The optimized liposomes formulation (RL5) was spherical with a vesicle size (VS) of 145.78 ± 9.9 nm, ζ potential (ZP) of 38.03 ± 9.12 mV, and encapsulation efficiency (EE) of 78.14 ± 8.04%. In vitro cytotoxicity studies in HepG2 cells demonstrated an improved anti-cancer activity of RL5 in comparison with free RS. These outcomes were supported by a cell uptake study in HepG2 cells, in which RL5 exhibited a higher uptake than free RS. Furthermore, confocal images of HepG2 cells after 3 and 5 h of incubation showed higher internalization of coumarin 6 (C6) loaded liposomes (CL) as compared to those of the free C6. Pharmacokinetic and pharmacodynamic (prophylactic and therapeutic treatment modalities) studies were performed in N-nitrosodiethylamine (NDEA-carcinogen) induced HCC in rats. Pharmacokinetic evaluation of RL5 demonstrated increased localization of RS in cancerous liver tissues by 3.2- and 2.2-fold increase in AUC and Cmax, respectively, when compared to those of the free RS group. A pharmacodynamic investigation revealed a significant reduction in hepatocyte nodules in RL5 treated animals when compared to those of free RS. Further, on treatment with RL5, HCC-bearing rats showed a significant decrease in the liver marker enzymes (alanine transaminase, alkaline phosphatase, aspartate transaminase, total bilirubin levels, γ-glutamyl transpeptidase, and α-fetoprotein), in comparison with that of the disease control group. Our findings were supported by histopathological analysis, and we were first to demonstrate that NDEA induced detrimental effect on rat livers was successfully reversed with the treatment of RL5 formulation. These results implied that delivery of RS loaded cationic liposomes substantially controlled the severity of HCC and that they can be considered as a promising nanocarrier in the management of HCC.
Collapse
Affiliation(s)
- Satveer Jagwani
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Nehru Nagar, Belagavi 590010, Karnataka, India.,Dr. Prabhakar Kore Basic Science Research Center, KLE Academy of Higher Education and Research, Nehru Nagar, Belagavi 590010, Karnataka, India
| | - Sunil Jalalpure
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Nehru Nagar, Belagavi 590010, Karnataka, India.,Dr. Prabhakar Kore Basic Science Research Center, KLE Academy of Higher Education and Research, Nehru Nagar, Belagavi 590010, Karnataka, India
| | - Dinesh Dhamecha
- Dr. Prabhakar Kore Basic Science Research Center, KLE Academy of Higher Education and Research, Nehru Nagar, Belagavi 590010, Karnataka, India
| | - Kiran Jadhav
- KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research, Nehru Nagar, Belagavi 590010, Karnataka, India
| | - Raghvendra Bohara
- Centre for Interdisciplinary Research, D. Y. Patil Education Society (Institution Deemed to be University), Line Bazar, Kasaba Bawada, Kolhapur, 416006, Maharashtra, India.,CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland, Upper New Castle, Galway, H91 W2TY, Ireland
| |
Collapse
|
35
|
Younis MA, Khalil IA, Harashima H. Gene Therapy for Hepatocellular Carcinoma: Highlighting the Journey from Theory to Clinical Applications. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Mahmoud A. Younis
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences Hokkaido University Kita‐12, Nishi‐6, Kita‐ku Sapporo 060‐0812 Japan
- Faculty of Pharmacy Assiut University Assiut 71526 Egypt
| | - Ikramy A. Khalil
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences Hokkaido University Kita‐12, Nishi‐6, Kita‐ku Sapporo 060‐0812 Japan
- Faculty of Pharmacy Assiut University Assiut 71526 Egypt
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences Hokkaido University Kita‐12, Nishi‐6, Kita‐ku Sapporo 060‐0812 Japan
| |
Collapse
|
36
|
Meng Q, Cong H, Hu H, Xu FJ. Rational design and latest advances of codelivery systems for cancer therapy. Mater Today Bio 2020; 7:100056. [PMID: 32510051 PMCID: PMC7264083 DOI: 10.1016/j.mtbio.2020.100056] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/25/2020] [Accepted: 04/29/2020] [Indexed: 01/06/2023] Open
Abstract
Current treatments have limited effectiveness in treating tumors. The combination of multiple drugs or treatment strategies is widely studied to improve therapeutic effect and reduce adverse effects of cancer therapy. The codelivery system is the key to realize combined therapies. It is necessary to design and construct different codelivery systems in accordance with the variable structures and properties of cargoes and vectors. This review presented the typical design considerations about codelivery vectors for cancer therapy and described the current state of codelivery systems from two aspects: different types of vectors and collaborative treatment strategies. The commonly used loading methods of cargoes into the vectors, including physical and chemical processes, are discussed in detail. Finally, we outline the challenges and perspectives about the improvement of codelivery systems.
Collapse
Affiliation(s)
- Q.Y. Meng
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China
| | - H.L. Cong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China
| | - H. Hu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China
| | - F.-J. Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
37
|
Thorp EB, Boada C, Jarbath C, Luo X. Nanoparticle Platforms for Antigen-Specific Immune Tolerance. Front Immunol 2020; 11:945. [PMID: 32508829 PMCID: PMC7251028 DOI: 10.3389/fimmu.2020.00945] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Innovative approaches in nanoparticle design have facilitated the creation of new formulations of nanoparticles that are capable of selectively calibrating the immune response. These nanomaterials may be engineered to interact with specific cellular and molecular targets. Recent advancements in nanoparticle synthesis have enabled surface functionalization of particles that mimic the diversity of ligands on the cell surface. Platforms synthesized using these design principles, called "biomimetic" nanoparticles, have achieved increasingly sophisticated targeting specificity and cellular trafficking capabilities. This holds great promise for next generation therapies that seek to achieve immune tolerance. In this review, we discuss the importance of physical design parameters including size, shape, and biomimetic surface functionalization, on the biodistribution, safety and efficacy of biologic nanoparticles. We will also explore potential applications for immune tolerance for organ or stem cell transplantation.
Collapse
Affiliation(s)
- Edward B. Thorp
- Departments of Pathology & Pediatrics at Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Christian Boada
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, United States
| | - Clarens Jarbath
- Departments of Pathology & Pediatrics at Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, United States
- Duke Transplant Center, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
38
|
Chitosan/carboxymethylcellulose-stabilized poly(lactide-co-glycolide) particles as bio-based drug delivery carriers. Carbohydr Polym 2020; 242:116417. [PMID: 32564826 DOI: 10.1016/j.carbpol.2020.116417] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/20/2020] [Accepted: 05/01/2020] [Indexed: 12/15/2022]
Abstract
Poly(lactide-co-glycolide) (PLGA) colloidal particles stabilized by complexes of two oppositely charged polysaccharides, chitosan (cationic, CS) and sodium carboxymethylcellulose (anionic, NaCMC), were fabricated. Dichloromethane containing dissolved PLGA was first emulsified in an aqueous phase containing mixtures of CS and NaCMC. Evaporation of dichloromethane from the resulting emulsion led to CS/NaCMC-covered-PLGA particles. CS and NaCMC contents affected the short-term stability of PLGA particles and also their intrinsic characteristics. The particles displayed pH-dependent characteristic. Zeta potential varied from +54 to -50 mV when pH was varied from 3 to 10. CS/NaCMC-covered-PLGA particles showed colloidal stability, over a wider pH range as compared to CS-covered-PLGA particles. Curcumin, a model hydrophobic drug, was encapsulated into the particles up to 10 wt% of PLGA. The CS/NaCMC-covered-PLGA particles loaded with curcumin showed delayed release in mildly acidic conditions and faster release in neutral and basic conditions. Cytotoxicity experiments were carried out with human colorectal carcinoma cells.
Collapse
|
39
|
Chatterjee M, Jaiswal N, Hens A, Mahata N, Chanda N. Development of 6-Thioguanine conjugated PLGA nanoparticles through thioester bond formation: Benefits of electrospray mediated drug encapsulation and sustained release in cancer therapeutic applications. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 114:111029. [PMID: 32994006 DOI: 10.1016/j.msec.2020.111029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/26/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022]
Abstract
Polymeric nanoparticle-based successful delivery of hydrophobic drugs is highly desirable for its controlled and sustained release at the disease site, which is a challenge with the current synthesis methods. In the present study, an electrospray mediated facile one-step synthesis approach is explored in which a solution mixture of a hydrophobic drug, 6-thioguanine (Tg) and a biocompatible FDA approved polymer, Poly (d, l-lactide-co-glycolide) (PLGA) is injected in an applied electric field of suitable intensity to prepare drug encapsulated PLGA nanoparticles, PLGA-Tg with high yield. In order to explore the effect of external electric field on Tg loading and delivery applications, the nanoparticles are characterized using EDX, AFM, FESEM, TEM, FTIR, Raman, fluorescence, and mass spectroscopy techniques. The characterization studies indicate that the electric field mediated synthesis exhibits spherical nanoparticles with a homogenous core size distribution of ~60 nm, high encapsulation (~97.22%) and stable conjugation of Tg (via thioester linkages) with PLGA molecules in the presence of the applied electric field. The kinetic study demonstrates the 'anomalous diffusion' (non-Fickian diffusion) release mechanism in which Tg escapes from PLGA matrix with a slow, but steady diffusion rate and the sustained drug release profile continues for 60 days. To check the biological activity of the encapsulated Tg, in-vitro cell studies of the PLGA-Tg are performed on HeLa cells. The MTT assay shows significant cell death after 48 h of treatment, and the cellular internalization of the drug-loaded nanoparticles occurs through pinocytosis mediated uptake, which is established by the AFM analysis. The Raman and mass spectroscopy studies suggest that the PLGA-Tg nanoparticles are rapidly hydrolyzed inside cell cytoplasm to release Tg which initiates apoptosis-mediated cell death confirmed by as DNA fragmentation and membrane blebbing studies. The results clearly emphasize the benefits of electrospray based synthesis of polymeric nanodrug formulation through the formation of chemical bonds between polymer and drug molecules that could be easily implemented in the design and development of an effective nanotherapeutic platform with no typical 'burst effect,' prolonged release profile, and significant toxicity to the cancer cells.
Collapse
Affiliation(s)
- Manosree Chatterjee
- Material Processing and Microsystem Laboratory, CSIR - Central Mechanical Engineering Research Institute, Durgapur 713209, India; Department of Biotechnology, National Institute of Technology Durgapur, Durgapur 713209, India
| | - Namita Jaiswal
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur 713209, India
| | - Abhiram Hens
- Department of Chemical Engineering, National Institute of Technology Durgapur, Durgapur 713209, India
| | - Nibedita Mahata
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur 713209, India
| | - Nripen Chanda
- Material Processing and Microsystem Laboratory, CSIR - Central Mechanical Engineering Research Institute, Durgapur 713209, India.
| |
Collapse
|
40
|
Jefremow A, Neurath MF. Nanoparticles in Gastrooncology. Visc Med 2020; 36:88-94. [PMID: 32355665 PMCID: PMC7184848 DOI: 10.1159/000506908] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 02/28/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gastrointestinal malignancies have the greatest incidence and cancer-associated death rates worldwide. Routine therapeutic modalities include surgery, chemotherapy and radiation but they often fail to reach the goal of cancer-free survival. SUMMARY In the light of this urgent medical need for the treatment of GI tumors, nanotech-nology-based approaches, i.e. nanomedicine, promise new therapeutic options. Using nanoparticles instead of classically designed drugs, targeting anticancer agents directly to the tumor site may revolutionize both diagnostic and therapeutic tools thereby facilitating the identification and elimination of malignant cells. Importantly, diagnostic insight and therapeutic effects can be achieved simultaneously through the same nanoparticle. Additionally, a nanoparticle may be loaded with more than one agent, thereby further increasing the value and power of the nanotechnology approach in oncologic therapeutic concepts. Although most insight into mechanisms of nanomedicine has been gained from in vitro and preclinical in vivo models, few clinical trials have been conducted, and nanomedicine-based concepts are already part of standard treatment algorithms. However, despite substantial progress it remains a challenge to design nanoparticles that feature all desirable characteristics at the same time. KEY MESSAGES This review seeks to provide substantial insight into the current status of nanomedicine-based approaches employed for diagnostic and/or therapeutic purposes in the field of gastrointestinal cancers by highlighting achievements and pointing out unresolved issues that need to be further addressed by future research attempts.
Collapse
Affiliation(s)
| | - Markus F. Neurath
- Department of Internal Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
41
|
Abstract
Cancer immunotherapy has shown great potential as witnessed by an increasing number of immuno-oncology drug approvals in the past few years. Meanwhile, the field of nucleic acid therapeutics has made significant advancement. Nucleic acid therapeutics, such as plasmids, antisense oligonucleotides (ASO), small interfering RNA (siRNA) and microRNA, messenger RNA (mRNA), immunomodulatory DNA/RNA, and gene-editing guide RNA (gRNA) are attractive due to their versatile abilities to alter the expression of target endogenous genes or even synthetic genes, and modulate the immune responses. These abilities can play vital roles in the development of novel immunotherapy strategies. However, limited by the intrinsic physicochemical properties such as negative charges, hydrophilicity, as well as susceptibility to enzymatic degradation, the delivery of nucleic acid therapeutics faces multiple challenges. It is therefore pivotal to develop drug delivery systems that can carry, protect, and specifically deliver and release nucleic acid therapeutics to target tissues and cells. In this review, we attempted to summarize recent advances in nucleic acid therapeutics and the delivery systems for these therapeutics in cancer immunotherapy.
Collapse
Affiliation(s)
- Shurong Zhou
- Department of Pharmaceutics, Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology, Drug Discovery and Development (ISB3D), School of Pharmacy, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23219, USA
| | - Wenjie Chen
- Department of Pharmaceutics, Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology, Drug Discovery and Development (ISB3D), School of Pharmacy, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23219, USA
| | - Janet Cole
- Department of Pharmaceutics, Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology, Drug Discovery and Development (ISB3D), School of Pharmacy, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23219, USA
| | - Guizhi Zhu
- Department of Pharmaceutics, Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology, Drug Discovery and Development (ISB3D), School of Pharmacy, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23219, USA
| |
Collapse
|
42
|
Wischhusen JC, Chowdhury SM, Lee T, Wang H, Bachawal S, Devulapally R, Afjei R, Sukumar UK, Paulmurugan R. Ultrasound-mediated delivery of miRNA-122 and anti-miRNA-21 therapeutically immunomodulates murine hepatocellular carcinoma in vivo. J Control Release 2020; 321:272-284. [PMID: 32004588 DOI: 10.1016/j.jconrel.2020.01.051] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/20/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common cause of cancer-related mortality, and patients with HCC show poor response to currently available treatments, which demands new therapies. We recently developed a synthetic microRNA-based molecularly targeted therapy for improving HCC response to chemotherapy by eliminating drug resistance. We used ultrasound-targeted microbubble destruction (UTMD) to locally deliver microRNA-loaded nanoparticles to HCC. Since the immune microenvironment plays a crucial role in HCC disease development and response to treatment, and UTMD and microRNAs have the potential to interfere with the immune system, in this study we analyzed the immunomodulatory effects of UTMD and miRNAs in HCC. We used an immunocompetent syngeneic HCC mouse model for the study. We conducted cytokine profiling in tumor, lymph nodes, and serum of animals within the first 24 h of treatment to analyze changes in the level of pro- and antitumoral cytokines. The results showed: (1) Hepa1-6 syngeneic tumors expressed HCC-related cytokines, (2) UTMD-microRNA combination therapy triggered transient cytokine storms, and (3) delivery of microRNA-122 and anti-microRNA-21 affected the immune microenvironment by decreasing the level of GM-CSF in tumors while modulating protumoral IL-1α, IL-1β, IL-5, IL-6 and IL-17 and antitumoral IL-2 and IL-12 in tumor-proximal lymph nodes, and increasing IL-2 in the serum of tumor-bearing mice. Local delivery of targeted therapy by UTMD significantly reduced the concentration of IL-12 and IL-17 in lymph nodes of treated and contralateral tumors suggesting a systemic response. CONCLUSION: UTMD-mediated delivery of microRNA-122 and anti-microRNA-21 modulated the immune microenvironment of Hepa1-6 tumors at the level of cytokine expressions. Exploiting antitumoral immune effects could enhance the therapeutic efficacy of the proposed combination therapy for HCC.
Collapse
Affiliation(s)
- Jennifer C Wischhusen
- Apoptosis, Cancer and Development Laboratory, Centre de Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, 69008 Lyon, France; Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | | | - Taehwa Lee
- Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Huaijun Wang
- Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA.
| | - Sunitha Bachawal
- Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA.
| | - Rammohan Devulapally
- Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Rayhaneh Afjei
- Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA.
| | - Uday Kumar Sukumar
- Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA.
| | - Ramasamy Paulmurugan
- Department of Radiology, School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
43
|
Zuo X, Chen Z, Gao W, Zhang Y, Wang J, Wang J, Cao M, Cai J, Wu J, Wang X. M6A-mediated upregulation of LINC00958 increases lipogenesis and acts as a nanotherapeutic target in hepatocellular carcinoma. J Hematol Oncol 2020; 13:5. [PMID: 31915027 PMCID: PMC6951025 DOI: 10.1186/s13045-019-0839-x] [Citation(s) in RCA: 287] [Impact Index Per Article: 71.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/17/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) possess significant regulatory functions in multiple biological and pathological processes, especially in cancer. Dysregulated lncRNAs in hepatocellular carcinoma (HCC) and their therapeutic applications remain unclear. METHODS Differentially expressed lncRNA profile in HCC was constructed using TCGA data. LINC00958 expression level was examined in HCC cell lines and tissues. Univariate and multivariate analyses were performed to demonstrate the prognostic value of LINC00958. Loss-of-function and gain-of-function experiments were used to assess the effects of LINC00958 on cell proliferation, motility, and lipogenesis. Patient-derived xenograft model was established for in vivo experiments. RNA immunoprecipitation, dual luciferase reporter, biotin-labeled miRNA pull-down, fluorescence in situ hybridization, and RNA sequencing assays were performed to elucidate the underlying molecular mechanisms. We developed a PLGA-based nanoplatform encapsulating LINC00958 siRNA and evaluated its superiority for systemic administration. RESULTS We identified a lipogenesis-related lncRNA, LINC00958, whose expression was upregulated in HCC cell lines and tissues. High LINC00958 level independently predicted poor overall survival. Functional assays showed that LINC00958 aggravated HCC malignant phenotypes in vitro and in vivo. Mechanistically, LINC00958 sponged miR-3619-5p to upregulate hepatoma-derived growth factor (HDGF) expression, thereby facilitating HCC lipogenesis and progression. METTL3-mediated N6-methyladenosine modification led to LINC00958 upregulation through stabilizing its RNA transcript. A PLGA-based nanoplatform loaded with si-LINC00958 was developed for HCC systemic administration. This novel drug delivery system was controlled release, tumor targeting, safe, and presented satisfactory antitumor efficacy. CONCLUSIONS Our results delineate the clinical significance of LINC00958 in HCC and the regulatory mechanisms involved in HCC lipogenesis and progression, providing a novel prognostic indicator and promising nanotherapeutic target.
Collapse
Affiliation(s)
- Xueliang Zuo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China.,Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Liver Transplantation, Nanjing, 210029, China.,Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, 241001, China
| | - Zhiqiang Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Liver Transplantation, Nanjing, 210029, China
| | - Wen Gao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yao Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Liver Transplantation, Nanjing, 210029, China
| | - Jinguo Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Junfeng Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China
| | - Ming Cao
- Key Laboratory of Environment-Friendly Polymeric Materials of Anhui Province, School of Chemistry and Chemical Engineering, Anhui University, Hefei, 230601, China
| | - Juan Cai
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, 241001, China. .,Department of Oncology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, China. .,The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, 210008, China.
| | - Jindao Wu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Liver Transplantation, Nanjing, 210029, China. .,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, China.
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Liver Transplantation, Nanjing, 210029, China.
| |
Collapse
|
44
|
Zhang HT, Sun J, Yan Y, Cui SH, Wang H, Wang CH, Qiu C, Chen X, Ding JS, Qian HG, Wang JC, Zhang Q. Encapsulated microRNA by gemcitabine prodrug for cancer treatment. J Control Release 2019; 316:317-330. [PMID: 31733293 DOI: 10.1016/j.jconrel.2019.11.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/24/2019] [Accepted: 11/12/2019] [Indexed: 01/21/2023]
Abstract
Although microRNAs (miRNAs) function as the important tumor gene regulators, they still confront with many challenges in systemic delivery. Here, the amphiphilic gemcitabine-oleic acid prodrugs (GOA) binding miRNAs with hydrogen bond are assembled into nanoparticles (GOA/miR NPs) through hydrophobic interaction via denaturation-annealing processes and nano-precipitation technique. The non-cationic GOA/miR NPs with an average size of ~150 nm and a zeta potential of ~ - 15 mV exhibit a stable encapsulation of miRNAs with non-sequence selectivity. Either miR-122 or miR-34a encapsulated in the GOA/miR NPs is efficiently delivered into HepG2 cells and significantly downregulate the expression levels of target gene after lysosome escape and pH-responsive disassembly. Moreover, in vivo experiments demonstrate that the GOA/miR-122 NPs exhibit higher tumor accumulation. Compared to GOA micelles, GOA/miR-122 NPs displayed stronger tumor inhibition (73% regression) after intravenous injection in nude mice xenografted with HCC, along with rapid clearance in normal liver tissues. Furthermore, there is no significant influence on biochemical indicators and immune factors during the systematic administration of GOA/miR-122 NPs. The non-cationic GOA/miR NPs engineered by hydrogen bond interaction and hydrophobic forces show the enhanced synergistic antitumor efficacy and good biosafety, which will provide a potential nanomedcine for HCC treatment.
Collapse
Affiliation(s)
- Hai-Tao Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Jing Sun
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, China
| | - Yi Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, China
| | - Shi-He Cui
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, China
| | - Hao Wang
- School of Pharmaceutical Sciences, Ningxia Medical University, YinChuan, NingXia 750004, China
| | - Cheng-Han Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, China
| | - Chong Qiu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, China
| | - Xin Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, China
| | - Jin-Song Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Hong-Gang Qian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, China.
| | - Qiang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, XueYuan Rd 38, Haidian Dist, Beijing 100191, China
| |
Collapse
|
45
|
Sun Y, Ma W, Yang Y, He M, Li A, Bai L, Yu B, Yu Z. Cancer nanotechnology: Enhancing tumor cell response to chemotherapy for hepatocellular carcinoma therapy. Asian J Pharm Sci 2019; 14:581-594. [PMID: 32104485 PMCID: PMC7032247 DOI: 10.1016/j.ajps.2019.04.005] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 03/06/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest cancers due to its complexities, reoccurrence after surgical resection, metastasis and heterogeneity. In addition to sorafenib and lenvatinib for the treatment of HCC approved by FDA, various strategies including transarterial chemoembolization, radiotherapy, locoregional therapy and chemotherapy have been investigated in clinics. Recently, cancer nanotechnology has got great attention for the treatment of various cancers including HCC. Both passive and active targetings are progressing at a steady rate. Herein, we describe the lessons learned from pathogenesis of HCC and the understanding of targeted and non-targeted nanoparticles used for the delivery of small molecules, monoclonal antibodies, miRNAs and peptides. Exploring current efficacy is to enhance tumor cell response of chemotherapy. It highlights the opportunities and challenges faced by nanotechnologies in contemporary hepatocellular carcinoma therapy, where personalized medicine is increasingly becoming the mainstay. Overall objective of this review is to enhance our understanding in the design and development of nanotechnology for treatment of HCC.
Collapse
Affiliation(s)
- Yongbing Sun
- National Engineering Research Center for solid preparation technology of Chinese Medicines, Jiangxi University of Traditional Chinese Medicines, Nanchang 330006, China
| | - Wen Ma
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yuanyuan Yang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Mengxue He
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Aimin Li
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Lei Bai
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown 26506, USA
| | - Bin Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhiqiang Yu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
46
|
Hu Z, Qin J, Li T, Guo J. Thyroid cancer MR molecular imaging via SHP2-targeted nanoparticles. Int J Nanomedicine 2019; 14:7365-7373. [PMID: 31686812 PMCID: PMC6751334 DOI: 10.2147/ijn.s201358] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 08/15/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Molecular imaging has generated a great demand to develop targeted contrast agents for MR imaging. MATERIALS AND METHODS In this study, we synthesized Src homology 2-containing phosphotyrosine phosphatase 2 (SHP2)-targeted and polylactic-co-glycolic acid--based nanoparticles (NPs), which encapsulated perfluoropentane and being chelated with gadolinium (Gd3+) as an efficient molecular probe for targeting MR imaging on thyroid carcinoma. RESULTS These NPs displayed practical properties and favorable biocompatibility in vitro. Furthermore, they showed abilities to specifically target thyroid cancer and enhance MRI as a contrast agent in both in vitro and in vivo experiments. CONCLUSION This novel MR molecular imaging based on this SHP2-targeted contrast agent provides a useful and non-invasive method for the early detection of thyroid carcinoma.
Collapse
Affiliation(s)
- ZhongQian Hu
- Department of Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu210009, People’s Republic of China
| | - JiaLe Qin
- Department of Ultrasound, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310000, People’s Republic of China
| | - TianKuan Li
- Department of Ultrasound, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu210009, People’s Republic of China
| | - JinHe Guo
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu210009, People’s Republic of China
| |
Collapse
|
47
|
Nanomedicine as a putative approach for active targeting of hepatocellular carcinoma. Semin Cancer Biol 2019; 69:91-99. [PMID: 31421265 DOI: 10.1016/j.semcancer.2019.08.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 08/04/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022]
Abstract
The effectiveness of chemotherapy in hepatocellular carcinoma (HCC) is restricted by chemo-resistance and systemic side effects. To improve the efficacy and safety of chemotherapeutics in HCC management, scientists have attempted to deliver these drugs to malignant tissues using targeted carriers as nanoparticles (NPs). Among the three types of NPs targeting (active, passive, and stimuli-responsive), active targeting is the most commonly investigated in HCC treatment. Despite the observed promising results so far, clinical research on nanomedicine targeting for HCC treatment still faces many challenges.These include batch-to-batch physicochemical properties' variations, limiting large scale production and insufficient data on human and environmental toxicities. This review summarized the characteristics of different nanocarriers, ligands, targeted receptors on HCC cells and provided recommendations to overcome the challenges, facing this novel line of treatment for HCC.
Collapse
|
48
|
Du M, Ouyang Y, Meng F, Ma Q, Liu H, Zhuang Y, Pang M, Cai T, Cai Y. Nanotargeted agents: an emerging therapeutic strategy for breast cancer. Nanomedicine (Lond) 2019; 14:1771-1786. [PMID: 31298065 DOI: 10.2217/nnm-2018-0481] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Breast cancer is the most common female cancer worldwide and represents 12% of all cancer cases. Improvements in survival rates are largely attributed to improved screening and diagnosis. Conventional chemotherapy remains an important treatment option but it is beset with poor cell selectivity, serious side effects and resistance. Nanoparticle drug delivery systems bring promising opportunities to breast cancer treatment. They may improve chemotherapy by targeting drugs to tumors, generating high drug concentrations at tumors providing slow release of the drug, increased drug stability and concomitant reductions in side effects. The nanotechnology-based drug delivery approaches and the current research and application status of nano-targeted agents for breast cancer are discussed in this review to provide a basis for further study on targeted drug delivery systems.
Collapse
Affiliation(s)
- Manling Du
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Yong Ouyang
- Guangzhou Hospital of Integrated Traditional Chinese & Western Medicine, Guangzhou 510800, PR China
| | - Fansu Meng
- Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of TCM, Zhongshan, Guangdong 528400, PR China
| | - Qianqian Ma
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Hui Liu
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Yong Zhuang
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Mujuan Pang
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Tiange Cai
- College of Life Sciences, Liaoning University, Shenyang 110036, PR China
| | - Yu Cai
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China.,Cancer Research Institute of Jinan University, Guangzhou 510632, PR China
| |
Collapse
|
49
|
Ban E, Kwon TH, Kim A. Delivery of therapeutic miRNA using polymer-based formulation. Drug Deliv Transl Res 2019; 9:1043-1056. [DOI: 10.1007/s13346-019-00645-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
50
|
Bai Z, Wei J, Yu C, Han X, Qin X, Zhang C, Liao W, Li L, Huang W. Non-viral nanocarriers for intracellular delivery of microRNA therapeutics. J Mater Chem B 2019; 7:1209-1225. [DOI: 10.1039/c8tb02946f] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
MicroRNAs are small regulatory noncoding RNAs that regulate various biological processes. Herein, we will present the development of the strategies for intracellular miRNAs delivery, and specially focus on the rational designed routes, their mechanisms of action, as well as potential therapeutics used in the host cells orin vivostudies.
Collapse
Affiliation(s)
- Zhiman Bai
- School of Physics and Materials Science
- Anhui University
- Hefei 230601
- China
| | - Jing Wei
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM)
- Nanjing Tech University (NanjingTech)
- Nanjing 211816
- China
| | - Changmin Yu
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM)
- Nanjing Tech University (NanjingTech)
- Nanjing 211816
- China
| | - Xisi Han
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM)
- Nanjing Tech University (NanjingTech)
- Nanjing 211816
- China
| | - Xiaofei Qin
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM)
- Nanjing Tech University (NanjingTech)
- Nanjing 211816
- China
| | - Chengwu Zhang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM)
- Nanjing Tech University (NanjingTech)
- Nanjing 211816
- China
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene
- Guangdong Provincial Key Laboratory of Tropical Disease Research
- School of Public Health
- Southern Medical University
- Guangzhou 510515
| | - Lin Li
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM)
- Nanjing Tech University (NanjingTech)
- Nanjing 211816
- China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM)
- Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM)
- Nanjing Tech University (NanjingTech)
- Nanjing 211816
- China
| |
Collapse
|