1
|
Gupta T, Murtaza M. Advancing Targeted Therapies in Pancreatic Cancer: Leveraging Molecular Aberrations for Therapeutic Success. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025:S0079-6107(25)00016-1. [PMID: 39988056 DOI: 10.1016/j.pbiomolbio.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/03/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Pancreatic cancer is one of the most deadly with poor prognosis and overall survival rate due to the dense stroma in the tumors which often is challenging for the delivery of drug to penetrate deep inside the tumor bed and usually results in the progression of cancer. The conventional treatment such as chemotherapy, radiotherapy or surgery shows a minimal benefit in the survival due to the drug resistance, poor penetration, less radiosensitivity or recurrence of tumor. There is an urgent demand to develop molecular- level targeted therapies to achieve therapeutic efficacy in the pancreatic ductal adenocarcinoma (PDAC) patients. The precision oncology focuses on the unique attributes of the patient such as epigenome, proteome, genome, microbiome, lifestyle and diet habits which contributes to promote oncogenesis. The targeted therapy helps to target the mutated proteins responsible for controlling growth, division and metastasis of tumor in the cancer cells. It is very important to consider all the attributes of the patient to provide the suitable personalized treatment to avoid any severe side effects. In this review, we have laid emphasis on the precision medicine; the utmost priority is to improve the survival of cancer patients by targeting molecular mutations through transmembrane proteins, inhibitors, signaling pathways, immunotherapy, gene therapy or the use of nanocarriers for the delivery at the tumor site. It will become beneficial therapeutic window to be considered for the advanced stage pancreatic cancer patients to prolong their survival rate.
Collapse
Affiliation(s)
- Tanvi Gupta
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan.
| | - Mohd Murtaza
- Fermentation & Microbial Biotechnology Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180016, India.
| |
Collapse
|
2
|
Liang Y, He S, Liu Q, Liu T, Tan Y, Peng T, Huang C, Lu X, Zheng S. Vimentin, inversely correlating with infiltration of CD8 + T lymphocytes, promotes nuclear translocation of PD-L1 in esophageal squamous cell carcinoma. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119781. [PMID: 38901494 DOI: 10.1016/j.bbamcr.2024.119781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/04/2024] [Accepted: 06/05/2024] [Indexed: 06/22/2024]
Abstract
Vimentin has been considered a canonical marker of epithelial-mesenchymal transition (EMT) and is associated with tumor escape characterized by aberrant PD-L1 expression. However, whether there is a relationship between vimentin and PD-L1 in esophageal squamous cell carcinoma (ESCC) remains poorly understood. The immunological involvement of vimentin in ESCC was first analyzed by multiplex immunofluorescence staining in ESCC tissue microarray followed by a xenografted mouse model. In vivo, C57BL/6 mice were subcutaneously transplanted with AKR cells after stable silencing of vimentin. In vivo results showed that in addition to PD-L1 and PD-L2 expression, vimentin expression was inversely correlated with CD8+ T-cell infiltration. Mechanistically, vimentin can directly interact with PD-L1 and promote nuclear translocation of PD-L1 in AKR cells. In addition, SEMA6C, STC-2 and TRAILR2 were identified as cytokines modulated by vimentin. Blockade of STC-2 and TRAILR2 in co-culture with their own primary antibodies was shown to recruit more CD8+ T cells than controls. Together, these data strongly suggest targeting Vimenin to overcome the immune cycle in ESCC.
Collapse
Affiliation(s)
- Yan Liang
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, PR China
| | - Shuo He
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, PR China
| | - Qing Liu
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, PR China
| | - Tao Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, PR China
| | - Yiyi Tan
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, PR China
| | - Tianyuan Peng
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, PR China
| | - Conggai Huang
- Department of Pathology, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xiaomei Lu
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, PR China.
| | - Shutao Zheng
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, PR China.
| |
Collapse
|
3
|
Habibizadeh M, Lotfollahzadeh S, Mahdavi P, Mohammadi S, Tavallaei O. Nanoparticle-mediated gene delivery of TRAIL to resistant cancer cells: A review. Heliyon 2024; 10:e36057. [PMID: 39247341 PMCID: PMC11379606 DOI: 10.1016/j.heliyon.2024.e36057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), also known as APO2L, has emerged as a highly potential anticancer agent because of its capacity to effectively trigger apoptosis in tumor cells by specifically binding to either of its death receptors (DR4 or DR5) while having no adverse effects on normal cells. Nevertheless, its practical use has been hindered by its inefficient pharmacokinetics characteristics, the challenges involved in its administration and delivery to targeted cells, and the resistance exhibited by most cancer cells towards TRAIL. Gene therapy, as a promising approach would be able to potentially circumvent TRAIL-based cancer therapy challenges mainly through localized TRAIL expression and generating a bystander impact. Among different strategies, using nanoparticles in TRAIL gene delivery allows for precise targeting, and overcoming TRAIL resistance by combination therapy. In this review, we go over potential mechanisms by which cancer cells achieve resistance to TRAIL and provide an overview of different carriers for delivering of the TRAIL gene to resistant cancer cells, focusing on different types of nanoparticles utilized in this context. We will also explore the challenges, and investigate future perspectives of this nanomedicine approach for cancer therapy.
Collapse
Affiliation(s)
- Mina Habibizadeh
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shima Lotfollahzadeh
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parisa Mahdavi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soheila Mohammadi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Omid Tavallaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
4
|
Lin J, Wang X, Ni D, Chen Y, Chen C, Liu Y. Combinational Gene Therapy toward Cancer with Nanoplatform: Strategies and Principles. ACS MATERIALS AU 2023; 3:584-599. [PMID: 38089659 PMCID: PMC10636764 DOI: 10.1021/acsmaterialsau.3c00035] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 12/18/2024]
Abstract
Cancer remains a significant threat to human health. While numerous therapies have been developed to combat the disease, traditional treatments such as chemotherapy and radiotherapy are suboptimal and associated with significant side effects. Gene therapy is an emerging therapeutic approach that offers improved targeting and reduced side effects compared with traditional treatments. Using siRNA and other nucleic acid-based drugs in cancer treatment has generated significant interest among researchers. Nanocarriers, such as liposomes, can effectively deliver these agents to tumor sites. However, gene therapy alone is often insufficient to eradicate tumors, and there is a risk of recurrence. Therefore, combining gene therapy with other therapies using nanocarriers, such as phototherapy and magnetic hyperthermia therapy, can lead to synergistic therapeutic effects through different mechanisms. In this review, we summarize various ways in which gene therapy can be combined with other therapies and highlight the role of nanoplatforms in mediating these combined therapies, which would inspire novel design ideas toward combination therapies. Additionally, bottlenecks and barriers to gene therapy should be addressed in the near future to achieve better clinical efficacy.
Collapse
Affiliation(s)
- Jinhui Lin
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University
of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Xinlian Wang
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University
of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Dongqi Ni
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University
of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Yandong Chen
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
| | - Chunying Chen
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University
of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Ying Liu
- CAS
Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
| |
Collapse
|
5
|
Zhao Y, Bo T, Wang C, Yao D, Pan C, Xu W, Zhou H, Li M, Zhang S. Superior TRAIL gene expression and cancer cell apoptosis mediated by highly branched-linear poly(β-amino ester)s. J Nanobiotechnology 2023; 21:394. [PMID: 37898777 PMCID: PMC10612241 DOI: 10.1186/s12951-023-02169-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023] Open
Abstract
Extensive efforts have been dedicated to enhancing the expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in cancer cells for the development of effective cancer treatments. However, highly safe and efficient delivery of TRAIL gene remains a significant challenge, especially using cationic polymers. Here, a series of highly branched-linear poly(β-amino ester)s (H-LPAEs) are developed through a unique oligomer branching strategy. H-LPAEs exhibit a more uniform distribution of linear segments and branching units, leading to excellent DNA condensation and favorable physicochemical properties of H-LPAE/DNA polyplexes. In SW1353 and BMSC cells, the optimized H-LPAEs, H-LPAEB4-S5-TMPTA, achieves superior gene transfection efficiency of 58.0% and 33.4%, which were 2.5-fold and 2.0-fold higher than that of the leading commercial gene transfection reagent, Lipofectamine 3000. Excitingly, H-LPAEB4-S5-TMPTA mediated 56.7% and 28.1% cell apoptosis in HepG2 cells and HeLa cells highlighting its potential application in cancer gene therapy. In addition, locally administered H-LPAEB4-S5-TMPTA delivered TRAIL DNA to HepG2 xenograft tumors and inhibited tumor growth in vivo. This study not only proposes a novel strategy for synthesizing poly(β-amino ester)s with a unique branched-linear topology but also identifies a promising candidate for highly efficient TRAIL gene transfection.
Collapse
Affiliation(s)
- Yitong Zhao
- School of Medicine, Anhui University of Science and Technology, 232000, Huainan, Anhui, China
| | - Tao Bo
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China
| | - Chenfei Wang
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China.
| | - Dingjin Yao
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Chaolan Pan
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Weiyi Xu
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China
| | - Hao Zhou
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Ming Li
- Department of Dermatology, Children's Hospital of Fudan University, National Children's Medical Center, 201102, Shanghai, China.
| | - Si Zhang
- School of Medicine, Anhui University of Science and Technology, 232000, Huainan, Anhui, China.
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
6
|
Pervaiz A, Saleem T, Kanwal K, Raza SM, Iqbal S, Zepp M, Georges RB, Berger MR. Expression profiling of anticancer genes in colorectal cancer patients and their in vitro induction by riproximin, a ribosomal inactivating plant protein. J Cancer Res Clin Oncol 2023; 149:4825-4837. [PMID: 36251065 DOI: 10.1007/s00432-022-04410-6] [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: 09/05/2022] [Accepted: 10/06/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Ectopic expression of anticancer genes (ACGs) imposes antineoplastic effects on transformed cells. Clinically, reduced expression of these genes has been linked with poor prognosis, metastasis and chemo/radiotherapy resistance in cancers. Identifying expression pattern of ACGs is crucial to establish their prognostic and therapeutic relevance in colorectal cancer (CRC). In addition to the clinical perspective, naturally occurring compounds can be explored in parallel for inducing ACGs to achieve cancer cell-specific death. METHODOLOGY Expression profiles of three ACGs (NOXA, PAR-4, TRAIL) were identified via real-time PCR in CRC clinical isolates. Time lapse-based expression modifications in ACGs were studied in a CRC liver metastasis animal model using microarray methodology. Effects of a purified plant protein (riproximin) on selected ACGs were identified in three primary and metastatic CRC cell lines by real-time PCR. Lastly, importance of the ACGs in a cellular environment was highlighted via bioinformatic analysis. RESULTS ACGs (except NOXA) were persistently downregulated in clinical isolates when comparing the overall mean expression values with normal mucosa levels. In vivo studies showed a prominent inhibition of NOXA and PAR-4 genes in implanted CRC cells during rat liver colonization. TRAIL showed deviation from this theme while showing marked induction during the early period of liver colonization (days 3 and 6 after CRC cell implantation). Riproximin exhibited substantial potential of inducing ACGs at transcriptome levels in selected CRC cell lines. Bioinformatic analysis showed that vital molecular/functional aspects of a cell are associated with the presence of ACGs. CONCLUSION ACGs are downregulated in primary and metastatic phase of CRC. Riproximin effectively induces ACGs in CRC cells and can be exploited for clinical investigations over time.
Collapse
Affiliation(s)
- Asim Pervaiz
- Institute of Biomedical and Allied Health Sciences, University of Health Sciences, Lahore, Pakistan.
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Talha Saleem
- Institute of Biomedical and Allied Health Sciences, University of Health Sciences, Lahore, Pakistan
- Department of Allied Health Sciences, Superior University, Lahore, Pakistan
| | - Kinzah Kanwal
- Institute of Biomedical and Allied Health Sciences, University of Health Sciences, Lahore, Pakistan
| | - Syed Mohsin Raza
- Institute of Biomedical and Allied Health Sciences, University of Health Sciences, Lahore, Pakistan
| | - Sana Iqbal
- Human Genetics and Molecular Biology Department, University of Health Sciences, Lahore, Pakistan
| | - Michael Zepp
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Immundiagnostik, Bensheim, Germany
| | - Rania B Georges
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Coordination Centre for Clinical Trials, University Hospital, Heidelberg, Germany
| | - Martin R Berger
- Toxicology and Chemotherapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Immundiagnostik, Bensheim, Germany
| |
Collapse
|
7
|
Mukherjee AG, Wanjari UR, Gopalakrishnan AV, Bradu P, Biswas A, Ganesan R, Renu K, Dey A, Vellingiri B, El Allali A, Alsamman AM, Zayed H, George Priya Doss C. Evolving strategies and application of proteins and peptide therapeutics in cancer treatment. Biomed Pharmacother 2023; 163:114832. [PMID: 37150032 DOI: 10.1016/j.biopha.2023.114832] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/18/2023] [Accepted: 04/30/2023] [Indexed: 05/09/2023] Open
Abstract
Several proteins and peptides have therapeutic potential and can be used for cancer therapy. By binding to cell surface receptors and other indicators uniquely linked with or overexpressed on tumors compared to healthy tissue, protein biologics enhance the active targeting of cancer cells, as opposed to the passive targeting of cells by conventional small-molecule chemotherapeutics. This study focuses on peptide medications that exist to slow or stop tumor growth and the spread of cancer, demonstrating the therapeutic potential of peptides in cancer treatment. As an alternative to standard chemotherapy, peptides that selectively kill cancer cells while sparing healthy tissue are developing. A mountain of clinical evidence supports the efficacy of peptide-based cancer vaccines. Since a single treatment technique may not be sufficient to produce favourable results in the fight against cancer, combination therapy is emerging as an effective option to generate synergistic benefits. One example of this new area is the use of anticancer peptides in combination with nonpeptidic cytotoxic drugs or the combination of immunotherapy with conventional therapies like radiation and chemotherapy. This review focuses on the different natural and synthetic peptides obtained and researched. Discoveries, manufacture, and modifications of peptide drugs, as well as their contemporary applications, are summarized in this review. We also discuss the benefits and difficulties of potential advances in therapeutic peptides.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India.
| | - Pragya Bradu
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Antara Biswas
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, South Korea
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077 Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal 700073, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda 151401, Punjab, India
| | - Achraf El Allali
- African Genome Center, Mohammed VI Polytechnic University, Ben Guerir, Morocco.
| | - Alsamman M Alsamman
- Department of Genome Mapping, Molecular Genetics, and Genome Mapping Laboratory, Agricultural Genetic Engineering Research Institute, Giza, Egypt
| | - Hatem Zayed
- Department of Biomedical Sciences College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - C George Priya Doss
- Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| |
Collapse
|
8
|
Yagolovich AV, Gasparian ME, Dolgikh DA. Recent Advances in the Development of Nanodelivery Systems Targeting the TRAIL Death Receptor Pathway. Pharmaceutics 2023; 15:pharmaceutics15020515. [PMID: 36839837 PMCID: PMC9961178 DOI: 10.3390/pharmaceutics15020515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/20/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
The TRAIL (TNF-related apoptosis-inducing ligand) apoptotic pathway is extensively exploited in the development of targeted antitumor therapy due to TRAIL specificity towards its cognate receptors, namely death receptors DR4 and DR5. Although therapies targeting the TRAIL pathway have encountered many obstacles in attempts at clinical implementation for cancer treatment, the unique features of the TRAIL signaling pathway continue to attract the attention of researchers. Special attention is paid to the design of novel nanoscaled delivery systems, primarily aimed at increasing the valency of the ligand for improved death receptor clustering that enhances apoptotic signaling. Optionally, complex nanoformulations can allow the encapsulation of several therapeutic molecules for a combined synergistic effect, for example, chemotherapeutic agents or photosensitizers. Scaffolds for the developed nanodelivery systems are fabricated by a wide range of conventional clinically approved materials and innovative ones, including metals, carbon, lipids, polymers, nanogels, protein nanocages, virus-based nanoparticles, dendrimers, DNA origami nanostructures, and their complex combinations. Most nanotherapeutics targeting the TRAIL pathway are aimed at tumor therapy and theranostics. However, given the wide spectrum of action of TRAIL due to its natural role in immune system homeostasis, other therapeutic areas are also involved, such as liver fibrosis, rheumatoid arthritis, Alzheimer's disease, and inflammatory diseases caused by bacterial infections. This review summarizes the recent innovative developments in the design of nanodelivery systems modified with TRAIL pathway-targeting ligands.
Collapse
Affiliation(s)
- Anne V. Yagolovich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Correspondence:
| | - Marine E. Gasparian
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | - Dmitry A. Dolgikh
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
9
|
TRAIL/S-layer/graphene quantum dot nanohybrid enhanced stability and anticancer activity of TRAIL on colon cancer cells. Sci Rep 2022; 12:5851. [PMID: 35393438 PMCID: PMC8991220 DOI: 10.1038/s41598-022-09660-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 03/21/2022] [Indexed: 12/14/2022] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL), known as a cytokine of the TNF superfamily, is considered a promising antitumor agent due to its ability to selectively induce apoptosis in a wide variety of cancer cells. However, failure of its successful translation into clinic has led to development of nano-based platforms aiming to improve TRAIL therapeutic efficacy. In this regard, we fabricated a novel TRAIL-S-layer fusion protein (S-TRAIL) conjugated with graphene quantum dots (GQDs) to benefit both the self-assembly of S-layer proteins, which leads to elevated TRAIL functional stability, and unique optical properties of GQDs. Noncovalent conjugation of biocompatible GQDs and soluble fusion protein was verified via UV–visible and fluorescence spectroscopy, size and ζ-potential measurements and transmission electron microscopy. The potential anticancer efficacy of the nanohybrid system on intrinsically resistant cells to TRAIL (HT-29 human colon carcinoma cells) was investigated by MTT assay and flow cytometry, which indicated about 80% apoptosis in cancer cells. These results highlight the potential of TRAIL as a therapeutic protein that can be extensively improved by taking advantage of nanotechnology and introduce S-TRAIL/GQD complex as a promising nanohybrid system in cancer treatment.
Collapse
|
10
|
Peng H, Guo X, He J, Duan C, Yang M, Zhang X, Zhang L, Fu R, Wang B, Wang D, Chen H, Xie M, Feng P, Dai L, Tang X, Luo J. Intracranial delivery of synthetic mRNA to suppress glioblastoma. Mol Ther Oncolytics 2022; 24:160-170. [PMID: 35024442 PMCID: PMC8724946 DOI: 10.1016/j.omto.2021.12.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 12/07/2021] [Indexed: 11/27/2022] Open
Abstract
Owing to messenger RNA's unique biological advantages, it has received increasing attention to be used as a therapeutic, known as mRNA-based gene therapy. It is critical to have an ideal strategy of mRNA gene therapy for glioma, which grows in a special environment. In the present study, we screened out a safe and efficient transfection reagent for intracranial delivery of synthetic mRNA in mouse brain. First, in order to analyze the effect of different transfection reagents on the intracranial delivery of mRNA, the synthetic luciferase mRNA was wrapped with two different transfection reagents and microinjected into the brain at the fixed point. The expression status of delivered mRNA was monitored by a small animal imaging system. The possible reagent-induced biological toxicity was evaluated by behavioral and blood biochemical measurements. Then, to test the therapeutic effect of our intracranial delivery mRNA model on glioma, synthetic modified tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) mRNA was used as an example of therapeutic application. This model demonstrated that synthetic mRNA could be successfully delivered into the brain using commercially available transfection reagents, and TransIT-mRNA showed better results than in vivo-jetPEI kit. This model can be applied in precise targeting and personalized gene therapy of glioma.
Collapse
Affiliation(s)
- Hao Peng
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan 442000, Hubei, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Xingrong Guo
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Jinjuan He
- Department of Respiratory and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Chao Duan
- Brain Research Institute, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Minghuan Yang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan 442000, Hubei, China
| | - Xianghua Zhang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan 442000, Hubei, China
| | - Li Zhang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan 442000, Hubei, China
| | - Rui Fu
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan 442000, Hubei, China
| | - Bin Wang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan 442000, Hubei, China
| | - Dekang Wang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan 442000, Hubei, China
| | - Hu Chen
- Medical Imaging Center, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Mengying Xie
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan 442000, Hubei, China
| | - Ping Feng
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan 442000, Hubei, China
| | - Longjun Dai
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan 442000, Hubei, China
| | - Xiangjun Tang
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan 442000, Hubei, China
| | - Jie Luo
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan 442000, Hubei, China
| |
Collapse
|
11
|
Karim MR, Naryan Panda S, Barman A, Sarkar I. Strain and crystallite size controlled ordering of Heusler nanoparticles having high heating rate for magneto-thermal application. NANOTECHNOLOGY 2022; 33:235701. [PMID: 35189616 DOI: 10.1088/1361-6528/ac56f7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/21/2022] [Indexed: 06/14/2023]
Abstract
Heusler compound nanoparticles with good structural ordering need to be investigated as a potential material class for magneto-thermal applications requiring heat generation in presence of an oscillating magnetic field. Here, we report an important finding of a structural parameter related to the product of the strain and the coherent crystallite size, that can be used to efficiently control the structural ordering and the magnetic property of the Heulser compound nanoparticles. The optimization of this product parameter is found to enhance both the structural ordering and magnetic transition temperature in Co2FeSn Heusler nanoparticles. Furthermore, using magnetic hyperthermia measurements we demonstrate the possibility of heat generation using Heusler compound nanoparticles comparable to that of conventional magnetic nanoparticles. This shall lead to the development of Heulser compounds for similar applications.
Collapse
Affiliation(s)
- Md Rejaul Karim
- Institute of Nano Science and Technology, Sector 81, Sahibzada Ajit Singh Nagar, Punjab 140306, India
| | - Surya Naryan Panda
- Department of Condensed Matter Physics and Material Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata 700106, India
| | - Anjan Barman
- Department of Condensed Matter Physics and Material Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata 700106, India
| | - Indranil Sarkar
- Institute of Nano Science and Technology, Sector 81, Sahibzada Ajit Singh Nagar, Punjab 140306, India
| |
Collapse
|
12
|
Roacho-Pérez JA, Garza-Treviño EN, Delgado-Gonzalez P, G-Buentello Z, Delgado-Gallegos JL, Chapa-Gonzalez C, Sánchez-Domínguez M, Sánchez-Domínguez CN, Islas JF. Target Nanoparticles against Pancreatic Cancer: Fewer Side Effects in Therapy. Life (Basel) 2021; 11:1187. [PMID: 34833063 PMCID: PMC8620707 DOI: 10.3390/life11111187] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 10/28/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is the most common lethal tumor in America. This lethality is related to limited treatment options. Conventional treatments involve the non-specific use of chemotherapeutical agents such as 5-FU, capecitabine, gemcitabine, paclitaxel, cisplatin, oxaliplatin, or irinotecan, which produce several side effects. This review focuses on the use of targeted nanoparticles, such as metallic nanoparticles, polymeric nanoparticles, liposomes, micelles, and carbon nanotubes as an alternative to standard treatment for pancreatic cancer. The principal objective of nanoparticles is reduction of the side effects that conventional treatments produce, mostly because of their non-specificity. Several molecular markers of pancreatic cancer cells have been studied to target nanoparticles and improve current treatment. Therefore, properly functionalized nanoparticles with specific aptamers or antibodies can be used to recognize pancreatic cancer cells. Once cancer is recognized, these nanoparticles can attack the tumor by drug delivery, gene therapy, or hyperthermia.
Collapse
Affiliation(s)
- Jorge A. Roacho-Pérez
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.D.-G.); (Z.G.-B.); (J.L.D.-G.); (C.N.S.-D.)
| | - Elsa N. Garza-Treviño
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.D.-G.); (Z.G.-B.); (J.L.D.-G.); (C.N.S.-D.)
| | - Paulina Delgado-Gonzalez
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.D.-G.); (Z.G.-B.); (J.L.D.-G.); (C.N.S.-D.)
| | - Zuca G-Buentello
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.D.-G.); (Z.G.-B.); (J.L.D.-G.); (C.N.S.-D.)
| | - Juan Luis Delgado-Gallegos
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.D.-G.); (Z.G.-B.); (J.L.D.-G.); (C.N.S.-D.)
| | - Christian Chapa-Gonzalez
- Instituto de Ingeniería y Tecnología, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez 32310, Mexico;
| | - Margarita Sánchez-Domínguez
- Grupo de Química Coloidal e Interfacial Aplicada a Nanomateriales y Formulaciones, Centro de Investigación en Materiales Avanzados, S.C. (CIMAV, S.C.), Unidad Monterrey, Apodaca 66628, Mexico;
| | - Celia N. Sánchez-Domínguez
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.D.-G.); (Z.G.-B.); (J.L.D.-G.); (C.N.S.-D.)
| | - Jose Francisco Islas
- Departamento de Bioquímica y Medicina Molecular, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey 64460, Mexico; (J.A.R.-P.); (E.N.G.-T.); (P.D.-G.); (Z.G.-B.); (J.L.D.-G.); (C.N.S.-D.)
| |
Collapse
|
13
|
Yang X, Xia X, Xia XX, Sun Z, Yan D. Improving Targeted Delivery and Antitumor Efficacy with Engineered Tumor Necrosis Factor-Related Apoptosis Ligand-Affibody Fusion Protein. Mol Pharm 2021; 18:3854-3861. [PMID: 34543035 DOI: 10.1021/acs.molpharmaceut.1c00483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Tumor necrosis factor-related apoptosis ligand (TRAIL) is a promising protein candidate for selective apoptosis of a variety of cancer cells. However, the short half-life and a lack of targeted delivery are major obstacles for its application in cancer therapy. Here, we propose a simple strategy to solve the targeting problem by genetically fusing an anti-HER2 affibody to the C-terminus of the TRAIL. The fusion protein TRAIL-affibody was produced as a soluble form with high yield in recombinant Escherichia coli. In vitro studies proved that the affibody domain promoted the cellular uptake of the fusion protein in the HER2 overexpressed SKOV-3 cells and improved its apoptosis-inducing ability. In addition, the fusion protein exhibited higher accumulation at the tumor site and greater antitumor effect than those of TRAIL in vivo, indicating that the affibody promoted the tumor homing of the TRAIL and then improved the therapeutic efficacy. Importantly, repeated injection of high-dose TRAIL-affibody showed no obvious toxicity in mice. These results demonstrated that the engineered TRAIL-affibody is promising to be a highly tumor-specific and targeted cancer therapeutic agent.
Collapse
Affiliation(s)
- Xiaoyuan Yang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xuelin Xia
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xiao-Xia Xia
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Zhao Sun
- Shandong Luning Pharmaceutical Co. Ltd., Guangrao County, Shandong Province 257336, People's Republic of China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| |
Collapse
|
14
|
Yagolovich A, Kuskov A, Kulikov P, Kurbanova L, Bagrov D, Artykov A, Gasparian M, Sizova S, Oleinikov V, Gileva A, Kirpichnikov M, Dolgikh D, Markvicheva E. Amphiphilic Poly( N-vinylpyrrolidone) Nanoparticles Conjugated with DR5-Specific Antitumor Cytokine DR5-B for Targeted Delivery to Cancer Cells. Pharmaceutics 2021; 13:1413. [PMID: 34575490 PMCID: PMC8464842 DOI: 10.3390/pharmaceutics13091413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 11/17/2022] Open
Abstract
Nanoparticles based on the biocompatible amphiphilic poly(N-vinylpyrrolidone) (Amph-PVP) derivatives are promising for drug delivery. Amph-PVPs self-aggregate in aqueous solutions with the formation of micellar nanoscaled structures. Amph-PVP nanoparticles are able to immobilize therapeutic molecules under mild conditions. As is well known, many efforts have been made to exploit the DR5-dependent apoptosis induction for cancer treatment. The aim of the study was to fabricate Amph-PVP-based nanoparticles covalently conjugated with antitumor DR5-specific TRAIL (Tumor necrosis factor-related apoptosis-inducing ligand) variant DR5-B and to evaluate their in vitro cytotoxicity in 3D tumor spheroids. The Amph-PVP nanoparticles were obtained from a 1:1 mixture of unmodified and maleimide-modified polymeric chains, while DR5-B protein was modified by cysteine residue at the N-end for covalent conjugation with Amph-PVP. The nanoparticles were found to enhance cytotoxicity effects compared to those of free DR5-B in both 2D (monolayer culture) and 3D (tumor spheroids) in vitro models. The cytotoxicity of the nanoparticles was investigated in human cell lines, namely breast adenocarcinoma MCF-7 and colorectal carcinomas HCT116 and HT29. Notably, DR5-B conjugation with Amph-PVP nanoparticles sensitized resistant multicellular tumor spheroids from MCF-7 and HT29 cells. Taking into account the nanoparticles loading ability with a wide range of low-molecular-weight antitumor chemotherapeutics into hydrophobic core and feasibility of conjugation with hydrophilic therapeutic molecules by click chemistry, we suggest further development to obtain a versatile system for targeted drug delivery into tumor cells.
Collapse
Affiliation(s)
- Anne Yagolovich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.K.); (A.A.); (M.G.); (S.S.); (V.O.); (A.G.); (E.M.); (M.K.); (D.D.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Andrey Kuskov
- D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia;
| | - Pavel Kulikov
- Federal State Budgetary Institution “Centre for Strategic Planning and Management of Biomedical Health Risks” of the Federal Medical Biological Agency, 119121 Moscow, Russia;
| | - Leily Kurbanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.K.); (A.A.); (M.G.); (S.S.); (V.O.); (A.G.); (E.M.); (M.K.); (D.D.)
| | - Dmitry Bagrov
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Artem Artykov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.K.); (A.A.); (M.G.); (S.S.); (V.O.); (A.G.); (E.M.); (M.K.); (D.D.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Marine Gasparian
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.K.); (A.A.); (M.G.); (S.S.); (V.O.); (A.G.); (E.M.); (M.K.); (D.D.)
| | - Svetlana Sizova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.K.); (A.A.); (M.G.); (S.S.); (V.O.); (A.G.); (E.M.); (M.K.); (D.D.)
| | - Vladimir Oleinikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.K.); (A.A.); (M.G.); (S.S.); (V.O.); (A.G.); (E.M.); (M.K.); (D.D.)
| | - Anastasia Gileva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.K.); (A.A.); (M.G.); (S.S.); (V.O.); (A.G.); (E.M.); (M.K.); (D.D.)
| | - Mikhail Kirpichnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.K.); (A.A.); (M.G.); (S.S.); (V.O.); (A.G.); (E.M.); (M.K.); (D.D.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Dmitry Dolgikh
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.K.); (A.A.); (M.G.); (S.S.); (V.O.); (A.G.); (E.M.); (M.K.); (D.D.)
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Elena Markvicheva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (L.K.); (A.A.); (M.G.); (S.S.); (V.O.); (A.G.); (E.M.); (M.K.); (D.D.)
| |
Collapse
|
15
|
Calinescu AA, Kauss MC, Sultan Z, Al-Holou WN, O'Shea SK. Stem cells for the treatment of glioblastoma: a 20-year perspective. CNS Oncol 2021; 10:CNS73. [PMID: 34006134 PMCID: PMC8162173 DOI: 10.2217/cns-2020-0026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma, the deadliest form of primary brain tumor, remains a disease without cure. Treatment resistance is in large part attributed to limitations in the delivery and distribution of therapeutic agents. Over the last 20 years, numerous preclinical studies have demonstrated the feasibility and efficacy of stem cells as antiglioma agents, leading to the development of trials to test these therapies in the clinic. In this review we present and analyze these studies, discuss mechanisms underlying their beneficial effect and highlight experimental progress, limitations and the emergence of promising new therapeutic avenues. We hope to increase awareness of the advantages brought by stem cells for the treatment of glioblastoma and inspire further studies that will lead to accelerated implementation of effective therapies. Glioblastoma is the deadliest and most common form of brain tumor, for which there is no cure. It is very difficult to deliver medicine to the tumor cells, because they spread out widely into the normal brain, and local blood vessels represent a barrier that most medicines cannot cross. It was shown, in many studies over the last 20 years, that stem cells are attracted toward the tumor and that they can deliver many kinds of therapeutic agents directly to brain cancer cells and shrink the tumor. In this review we analyze these studies and present new discoveries that can be used to make stem cell therapies for glioblastoma more effective to prolong the life of patients with brain tumors.
Collapse
Affiliation(s)
| | - McKenzie C Kauss
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,College of Literature Science & Arts, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zain Sultan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA.,College of Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wajd N Al-Holou
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sue K O'Shea
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Belkahla H, Constantinescu AA, Gharbi T, Barbault F, Chevillot-Biraud A, Decorse P, Micheau O, Hémadi M, Ammar S. Grafting TRAIL through Either Amino or Carboxylic Groups onto Maghemite Nanoparticles: Influence on Pro-Apoptotic Efficiency. NANOMATERIALS 2021; 11:nano11020502. [PMID: 33671136 PMCID: PMC7922020 DOI: 10.3390/nano11020502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/11/2021] [Accepted: 02/16/2021] [Indexed: 11/16/2022]
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF cytokine superfamily. TRAIL is able to induce apoptosis through engagement of its death receptors DR4 and DR5 in a wide variety of tumor cells while sparing vital normal cells. This makes it a promising agent for cancer therapy. Here, we present two different ways of covalently grafting TRAIL onto maghemite nanoparticles (NPs): (a) by using carboxylic acid groups of the protein to graft it onto maghemite NPs previously functionalized with amino groups, and (b) by using the amino functions of the protein to graft it onto NPs functionalized with carboxylic acid groups. The two resulting nanovectors, NH-TRAIL@NPs-CO and CO-TRAIL@NPs-NH, were thoroughly characterized. Biological studies performed on human breast and lung carcinoma cells (MDA-MB-231 and H1703 cell lines) established these nanovectors are potential agents for cancer therapy. The pro-apoptotic effect is somewhat greater for CO-TRAIL@NPs-NH than NH-TRAIL@NPs-CO, as evidenced by viability studies and apoptosis analysis. A computational study indicated that regardless of whether TRAIL is attached to NPs through an acid or an amino group, DR4 recognition is not affected in either case.
Collapse
Affiliation(s)
- Hanene Belkahla
- Université de Paris, CNRS-UMR 7086, Interfaces, Traitements, Organisation et DYnamique des Systèmes (ITODYS), UFR de Chimie, 15 rue Jean-Antoine de Baïf, 75013 Paris, France; (H.B.); (F.B.); (A.C.-B.); (P.D.)
- Lipides Nutrition Cancer, INSERM-UMR 1231, Université de Bourgogne Franche-Comté, UFR Science de Santé, 7 Bd Jeanne d’Arc, 21000 Dijon, France; (A.A.C.); (O.M.)
- Nanomedicine, Imagery and Therapeutics, EA 4662, Université de Bourgogne Franche-Comté, UFR Sciences & Techniques, 16 Route de Gray, 25030 Besançon CEDEX, France;
| | - Andrei Alexandru Constantinescu
- Lipides Nutrition Cancer, INSERM-UMR 1231, Université de Bourgogne Franche-Comté, UFR Science de Santé, 7 Bd Jeanne d’Arc, 21000 Dijon, France; (A.A.C.); (O.M.)
| | - Tijani Gharbi
- Nanomedicine, Imagery and Therapeutics, EA 4662, Université de Bourgogne Franche-Comté, UFR Sciences & Techniques, 16 Route de Gray, 25030 Besançon CEDEX, France;
| | - Florent Barbault
- Université de Paris, CNRS-UMR 7086, Interfaces, Traitements, Organisation et DYnamique des Systèmes (ITODYS), UFR de Chimie, 15 rue Jean-Antoine de Baïf, 75013 Paris, France; (H.B.); (F.B.); (A.C.-B.); (P.D.)
| | - Alexandre Chevillot-Biraud
- Université de Paris, CNRS-UMR 7086, Interfaces, Traitements, Organisation et DYnamique des Systèmes (ITODYS), UFR de Chimie, 15 rue Jean-Antoine de Baïf, 75013 Paris, France; (H.B.); (F.B.); (A.C.-B.); (P.D.)
| | - Philippe Decorse
- Université de Paris, CNRS-UMR 7086, Interfaces, Traitements, Organisation et DYnamique des Systèmes (ITODYS), UFR de Chimie, 15 rue Jean-Antoine de Baïf, 75013 Paris, France; (H.B.); (F.B.); (A.C.-B.); (P.D.)
| | - Olivier Micheau
- Lipides Nutrition Cancer, INSERM-UMR 1231, Université de Bourgogne Franche-Comté, UFR Science de Santé, 7 Bd Jeanne d’Arc, 21000 Dijon, France; (A.A.C.); (O.M.)
| | - Miryana Hémadi
- Université de Paris, CNRS-UMR 7086, Interfaces, Traitements, Organisation et DYnamique des Systèmes (ITODYS), UFR de Chimie, 15 rue Jean-Antoine de Baïf, 75013 Paris, France; (H.B.); (F.B.); (A.C.-B.); (P.D.)
- Correspondence: (M.H.); (S.A.)
| | - Souad Ammar
- Université de Paris, CNRS-UMR 7086, Interfaces, Traitements, Organisation et DYnamique des Systèmes (ITODYS), UFR de Chimie, 15 rue Jean-Antoine de Baïf, 75013 Paris, France; (H.B.); (F.B.); (A.C.-B.); (P.D.)
- Correspondence: (M.H.); (S.A.)
| |
Collapse
|
17
|
Zheng Q, Zhang Q, Yu X, He Y, Guo W. FENDRR: A pivotal, cancer-related, long non-coding RNA. Biomed Pharmacother 2021; 137:111390. [PMID: 33761608 DOI: 10.1016/j.biopha.2021.111390] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/27/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have more than 200 nucleotides and do not encode proteins. Based on numerous studies, lncRNAs have emerged as new and crucial regulators of biological function and have been implicated in the pathogenesis of a variety of diseases, especially cancers. Specific lncRNAs have been identified as novel molecular biomarkers for cancer diagnosis, prognosis, and treatment efficacy. Fetal-lethal non-coding developmental regulatory RNA (FENDRR, also known as FOXF1-AS1) is a novel lncRNA that is located at chr3q13.31 and has four exons and 3099 nucleotides, and its genomic site is located at chr3q13.31. FENDRR is abnormally expressed in a variety of cancers and is significantly associated with different clinical characteristics. In addition, FENDRR has shown potential as a biomarker for cancer diagnosis, prognosis, and treatment. In this review, we summarize the current understanding of FENDRR and its mechanistic role in cancer progression. We also discuss recent insights into the clinical significance of FENDRR for cancer diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Qingyuan Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou 450052, China; Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou 450052, China
| | - Qiyao Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou 450052, China; Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou 450052, China
| | - Xiao Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou 450052, China; Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou 450052, China
| | - Yuting He
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou 450052, China; Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou 450052, China.
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou 450052, China; Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou 450052, China.
| |
Collapse
|
18
|
Wang J, Gu X, Ouyang Y, Chu L, Xu M, Wang K, Tong X. Engineering of Neutrophil Membrane Camouflaging Nanoparticles Realizes Targeted Drug Delivery for Amplified Antitumor Therapy. Int J Nanomedicine 2021; 16:1175-1187. [PMID: 33623381 PMCID: PMC7894798 DOI: 10.2147/ijn.s288636] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/23/2021] [Indexed: 12/11/2022] Open
Abstract
Purpose Although the neutrophil membrane (NM)-based nanoparticulate delivery system has exhibited rapid advances in tumor targeting stemmed from the inherited instinct, the antitumor effect requires further improvement due to inefficient cellular internalization in the absence of specific interactions between NM-coated nanoparticles and tumor cells. Methods Herein, we fabricated drug-paclitaxel loaded NM camouflaging nanoparticles (TNM-PN) modified with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), favorable for the cellular internalization. Results The results showed that TNM-PN exerted a significant cytotoxicity to tumor cells by TRAIL-mediated endocytosis and strong adhesion to inflamed endothelial cells in vitro. Due to TRAIL modification as well as the adhesive interactions between neutrophil and inflamed tumor vascular endothelial cells, tumors in TNM-PN group exhibited almost 2-fold higher fluorescence intensities than that of NM camouflaging nanoparticles and 3-fold higher than that of bare nanoparticles, respectively. Significant tumor inhibition and survival rates of mice were achieved in TNM-PN group as a consequence of prolonged blood circulations to 48 h and preferential tumor accumulations, which was ascribed to targeting adhesion originated from NM to immune evasion and subsequent excellent cellular internalization. Conclusion The research unveiled a novel strategy of amplifying cellular internalization based on NM coating nanotechnology to boost antitumor efficacy.
Collapse
Affiliation(s)
- Jingshuai Wang
- Obstetrics and Gynecology Department, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| | - Xuemin Gu
- Obstetrics and Gynecology Department, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| | - Yiqin Ouyang
- Obstetrics and Gynecology Department, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| | - Lei Chu
- Obstetrics and Gynecology Department, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| | - Mengjiao Xu
- Obstetrics and Gynecology Department, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| | - Kun Wang
- Cancer Center, Shanghai East Hospital of Tongji University, Shanghai, People's Republic of China
| | - Xiaowen Tong
- Obstetrics and Gynecology Department, Tongji Hospital of Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
19
|
Liu S, Qiu J, He G, He W, Liu C, Cai D, Pan H. TRAIL promotes hepatocellular carcinoma apoptosis and inhibits proliferation and migration via interacting with IER3. Cancer Cell Int 2021; 21:63. [PMID: 33472635 PMCID: PMC7816514 DOI: 10.1186/s12935-020-01724-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/19/2020] [Indexed: 12/26/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can induce substantial cytotoxicity in tumor cells but rarely exert cytotoxic activity on non-transformed cells. In the present study, we therefore evaluated interactions between TRAIL and IER3 via co-immunoprecipitation and immunofluorescence analyses, leading us to determine that these two proteins were able to drive the apoptotic death of hepatocellular carcinoma (HCC) cells and to disrupt their proliferative and migratory abilities both in vitro and in vivo. From a mechanistic perspective, we determined that TRAIL and IER3 were capable of inhibiting Wnt/β-catenin signaling. Together, these results indicate that TRAIL can control the pathogenesis of HCC at least in part via interacting with IER3 to inhibit Wnt/β-catenin signaling, thus indicating that this TRAIL/IER3/β-catenin axis may be a viable therapeutic target in HCC patients.
Collapse
Affiliation(s)
- Shihai Liu
- Medical Animal Lab, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Jing Qiu
- Department of Stomatology, Qingdao Municipal Hospital, Qingdao, 266071, China
| | - Guifang He
- Medical Animal Lab, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Weitai He
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Changchang Liu
- Medical Animal Lab, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Duo Cai
- Medical Animal Lab, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Huazheng Pan
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
20
|
Singh D, Tewari M, Singh S, Narayan G. Revisiting the role of TRAIL/TRAIL-R in cancer biology and therapy. Future Oncol 2021; 17:581-596. [PMID: 33401962 DOI: 10.2217/fon-2020-0727] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL), a member of the TNF superfamily, can induce apoptosis in cancer cells, sparing normal cells when bound to its associated death receptors (DR4/DR5). This unique mechanism makes TRAIL a potential anticancer therapeutic agent. However, clinical trials of recombinant TRAIL protein and TRAIL receptor agonist monoclonal antibodies have shown disappointing results due to its short half-life, poor pharmacokinetics and the resistance of the cancer cells. This review summarizes TRAIL-induced apoptotic and survival pathways as well as mechanisms leading to apoptotic resistance. Recent development of methods to overcome cancer cell resistance to TRAIL-induced apoptosis, such as protein modification, combination therapy and TRAIL-based gene therapy, appear promising. We also discuss the challenges and opportunities in the development of TRAIL-based therapies for the treatment of human cancers.
Collapse
Affiliation(s)
- Deepika Singh
- Department of Molecular & Human Genetics, Cancer Genetics Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Mallika Tewari
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Sunita Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, 221005, India
| | - Gopeshwar Narayan
- Department of Molecular & Human Genetics, Cancer Genetics Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
21
|
Yoo JD, Bae SM, Seo J, Jeon IS, Vadevoo SMP, Kim SY, Kim IS, Lee B, Kim S. Designed ferritin nanocages displaying trimeric TRAIL and tumor-targeting peptides confer superior anti-tumor efficacy. Sci Rep 2020; 10:19997. [PMID: 33203916 PMCID: PMC7672110 DOI: 10.1038/s41598-020-77095-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 11/05/2020] [Indexed: 12/15/2022] Open
Abstract
TRAIL is considered a promising target for cancer therapy because it mediates activation of the extrinsic apoptosis pathway in a tumor-specific manner by binding to and trimerizing its functional receptors, DR4 or DR5. Although recombinant human TRAIL has shown high potency and specificity for killing cancer cells in preclinical studies, it has failed in multiple clinical trials for several reasons, including a very short half-life mainly caused by instability of the monomeric form of TRAIL and rapid renal clearance of the off-targeted TRAIL. To overcome such obstacles, we developed a TRAIL-active trimer nanocage (TRAIL-ATNC) that presents the TRAIL ligand in its trimer-like conformation by connecting it to a triple helix sequence that links to the threefold axis of the ferritin nanocage. We also ligated the tumor-targeting peptide, IL4rP, to TRAIL-ATNC to enhance tumor targeting. The developed TRAIL-ATNCIL4rP showed enhanced agonistic activity compared with monomeric TRAIL. The in vivo serum half-life of TRAIL-ATNCIL4rP was ~ 16-times longer than that of native TRAIL. As a consequence of these properties, TRAIL-ATNCIL4rP exhibited efficacy as an anti-tumor agent in vivo against xenograft breast cancer as well as orthotopic pancreatic cancer models, highlighting the promise of this system for development as novel therapeutics against cancer.
Collapse
Affiliation(s)
- Jae Do Yoo
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Sang Mun Bae
- PrismCDX, Inc., 593-16, Dongtangiheung-ro, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Junyoung Seo
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - In Seon Jeon
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Sri Murugan Poongkavithai Vadevoo
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Soyoun Kim
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
| |
Collapse
|
22
|
Qiu Y, Sun J, Qiu J, Chen G, Wang X, Mu Y, Li K, Wang W. Antitumor Activity of Cabazitaxel and MSC-TRAIL Derived Extracellular Vesicles in Drug-Resistant Oral Squamous Cell Carcinoma. Cancer Manag Res 2020; 12:10809-10820. [PMID: 33149686 PMCID: PMC7605918 DOI: 10.2147/cmar.s277324] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/03/2020] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) can induce apoptosis in a variety of cancer cells. However, drug resistance of tumor and short half-life seriously affects its clinical targeted therapy. Cabazitaxel (CTX) is a taxane drug, which can induce apoptosis or autophagy by inhibiting the phosphorylation of PI3K/Akt/mTOR and sensitive to some drug-resistant tumors. Therefore, we explored the possibility of developing a mesenchymal stem cell-derived exosomes (MSC-EXO) vector for oral squamous cell carcinoma (OSCC) to deliver CTX/TRAIL combinations. METHODS After ultracentrifugation and dialysis, CTX/TRAIL loaded exosomes transfected MSC (MSCT)-derived exosome (EXO) (MSCT-EXO/CTX) were isolated and purified. The expression of CD63, CD9 and TRAIL was detected by BCA to confirm the origin of EXO. High-performance liquid chromatography (HPLC) was used to determine the drug loading of VPF and draw the in vitro release profile. MTT assay, flow cytometry and Western blot were used to detect the antitumor effect of MSCT-EXO/CTX in vitro. Subsequently, the antitumor effect of MSCT-EXO/CTX in vivo was verified by mouse model. RESULTS The diameter of the membrane particles was about 60-150 nm. We have proved that the incorporation and release of CTX in MSCT-EXO can inhibit the activation of PI3K, Akt and mTOR, which is a possible synergistic mechanism of CTX. MSCT-EXO and CTX can induce the apoptosis of SCC25 tumor cells in a dose-dependent manner and exert a good synergistic effect in the proportion range of 10:1-5:1. The inherent activity of MSCT-EXO and the direct effect of MSCT-EXO/CTX on OSCC confirm that MSCT-EXO/CTX makes MSCT-EXO and CTX have an efficient synergistic effect and a highly effective pharmacological inhibition on cancer cells, as verified by the subsequent mouse model. MSCT-EXO/CTX showed the lowest relative tumor volume and the highest tumor inhibition rate (P<0.05) in vivo. CONCLUSION An MSCT-EXO-based CTX delivery system might be an effective anticancer method.
Collapse
Affiliation(s)
- Yongle Qiu
- Department of Stomatology, Fourth Affiliated Hospital, Hebei Medical University, Shijiazhuang, Hebei050017, People’s Republic of China
| | - Jieming Sun
- Department of Stomatology, Xianghe County People ‘S Hospital, Langfang, Hebei065400, People’s Republic of China
| | - Junping Qiu
- Department of Stomatology, Xianghe County People ‘S Hospital, Langfang, Hebei065400, People’s Republic of China
| | - Guoling Chen
- Department of Stomatology, Xianghe County People ‘S Hospital, Langfang, Hebei065400, People’s Republic of China
| | - Xiao Wang
- Department of Stomatology, Xianghe County People ‘S Hospital, Langfang, Hebei065400, People’s Republic of China
| | - Yaxu Mu
- Department of Stomatology, Xianghe County People ‘S Hospital, Langfang, Hebei065400, People’s Republic of China
| | - Kunshan Li
- Department of Stomatology, Fourth Affiliated Hospital, Hebei Medical University, Shijiazhuang, Hebei050017, People’s Republic of China
| | - Wenjing Wang
- Department of Stomatology, Fourth Affiliated Hospital, Hebei Medical University, Shijiazhuang, Hebei050017, People’s Republic of China
| |
Collapse
|
23
|
Li Z, Wang W, Meng L, Zhang Y, Zhang J, Li C, Wu Y, Feng F, Zhang Q. Identification and analysis of key lncRNAs in malignant-transformed BEAS-2B cells induced with coal tar pitch by microarray analysis. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 79:103376. [PMID: 32470693 DOI: 10.1016/j.etap.2020.103376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 06/11/2023]
Abstract
This study aims to explore the key and differentially expressed long non-coding RNAs (lncRNAs) and elucidates their possible mechanisms in malignant-transformed Human bronchial epithelial (BEAS-2B) cells induced by coal tar pitch extracts (CTPE). BEAS-2B cells were stimulated with 2.4 μg/ml CTPE, then passaged for three times which were named CTPE1 and then passaged until passage 30 (CTPE30). The results showed that cells of CTPE30 appeared abnormal morphology. Furthermore, migration, clonality and proliferation of cells in CTPE group were significantly increased compared with those in control groups. However, the apoptosis of cells in CTPE group was inhibited. A total of 569 differentially expressed mRNAs and 707 differentially expressed lncRNAs were screened out, among which four lncRNAs were validated and were consistent with the microarray results. 32 target genes were screened out by Co-expression network. The study suggests that differentially expressed lncRNAs may play a potential role in lung carcinogenesis.
Collapse
Affiliation(s)
- Zhongqiu Li
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Weiguang Wang
- Rizhao Center for Disease Control and Prevention, Rizhao, Shandong province, 276800, China
| | - Liya Meng
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Yaping Zhang
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Jiatong Zhang
- Hospital of Zhengzhou University, Zhengzhou, Henan province, China
| | - Chunyang Li
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Yongjun Wu
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China
| | - Feifei Feng
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China.
| | - Qiao Zhang
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, Henan province, 450001, China.
| |
Collapse
|
24
|
Li H, Zhao J, Wang A, Li Q, Cui W. Supramolecular assembly of protein-based nanoparticles based on tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) for cancer therapy. Colloids Surf A Physicochem Eng Asp 2020. [DOI: 10.1016/j.colsurfa.2020.124486] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Belkahla H, Mazarío E, Sangnier AP, Lomas JS, Gharbi T, Ammar S, Micheau O, Wilhelm C, Hémadi M. TRAIL acts synergistically with iron oxide nanocluster-mediated magneto- and photothermia. Theranostics 2019; 9:5924-5936. [PMID: 31534529 PMCID: PMC6735372 DOI: 10.7150/thno.36320] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 06/09/2019] [Indexed: 02/06/2023] Open
Abstract
Targeting TRAIL (Tumor necrosis factor (TNF)-Related Apoptosis-Inducing Ligand) receptors for cancer therapy remains challenging due to tumor cell resistance and poor preparations of TRAIL or its derivatives. Herein, to optimize its therapeutic use, TRAIL was grafted onto iron oxide nanoclusters (NCs) with the aim of increasing its pro-apoptotic potential through nanoparticle-mediated magnetic hyperthermia (MHT) or photothermia (PT). Methods: The nanovector, NC@TRAIL, was characterized in terms of size, grafting efficiency, and potential for MHT and PT. The therapeutic function was assessed on a TRAIL-resistant breast cancer cell line, MDA-MB-231, wild type (WT) or TRAIL-receptor-deficient (DKO), by combining complementary methylene blue assay and flow cytometry detection of apoptosis and necrosis. Results: Combined with MHT or PT under conditions of "moderate hyperthermia" at low concentrations, NC@TRAIL acts synergistically with the TRAIL receptor to increase the cell death rate beyond what can be explained by the mere global elevation of temperature. In contrast, all results are consistent with the idea that there are hotspots, close to the nanovector and, therefore, to the membrane receptor, which cause disruption of the cell membrane. Furthermore, nanovectors targeting other membrane receptors, unrelated to the TNF superfamily, were also found to cause tumor cell damage upon PT. Indeed, functionalization of NCs by transferrin (NC@Tf) or human serum albumin (NC@HSA) induces tumor cell killing when combined with PT, albeit less efficiently than NC@TRAIL. Conclusions: Given that magnetic nanoparticles can easily be functionalized with molecules or proteins recognizing membrane receptors, these results should pave the way to original remote-controlled antitumoral targeted thermal therapies.
Collapse
Affiliation(s)
- Hanene Belkahla
- Université de Paris, ITODYS, CNRS-UMR 7086, 15 rue J.-A. de Baïf, F-75013 Paris, France
- Nanomedicine, Imagery and Therapeutics, EA 4662, Université de Bourgogne Franche-Comté, UFR Sciences & Techniques, 16 Route de Gray, 25030 Besançon Cedex, France
- Lipides nutrition cancer, INSERM-UMR 1231, Université de Bourgogne Franche-Comté, UFR Science de Santé, 7 Bd Jeanne d'Arc, 21000 Dijon, France
| | - Eva Mazarío
- Université de Paris, ITODYS, CNRS-UMR 7086, 15 rue J.-A. de Baïf, F-75013 Paris, France
| | - Anouchka Plan Sangnier
- Laboratoire Matières et Systèmes Complexes, Université de Paris, CNRS-UMR 7057, 10 rue Alice Domon et Léonie Duquet, 75205 Paris Cedex 13, France
| | - John S. Lomas
- Université de Paris, ITODYS, CNRS-UMR 7086, 15 rue J.-A. de Baïf, F-75013 Paris, France
| | - Tijani Gharbi
- Nanomedicine, Imagery and Therapeutics, EA 4662, Université de Bourgogne Franche-Comté, UFR Sciences & Techniques, 16 Route de Gray, 25030 Besançon Cedex, France
| | - Souad Ammar
- Université de Paris, ITODYS, CNRS-UMR 7086, 15 rue J.-A. de Baïf, F-75013 Paris, France
| | - Olivier Micheau
- Lipides nutrition cancer, INSERM-UMR 1231, Université de Bourgogne Franche-Comté, UFR Science de Santé, 7 Bd Jeanne d'Arc, 21000 Dijon, France
| | - Claire Wilhelm
- Laboratoire Matières et Systèmes Complexes, Université de Paris, CNRS-UMR 7057, 10 rue Alice Domon et Léonie Duquet, 75205 Paris Cedex 13, France
| | - Miryana Hémadi
- Université de Paris, ITODYS, CNRS-UMR 7086, 15 rue J.-A. de Baïf, F-75013 Paris, France
| |
Collapse
|
26
|
Hong S, Ding P, Luo Y, Gao T, Zhang Y, Pei R. Aptamer-integrated α-Gal liposomes as bispecific agents to trigger immune response for killing tumor cells. J Biomed Mater Res A 2019; 107:1176-1183. [PMID: 30650243 DOI: 10.1002/jbm.a.36609] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/03/2018] [Accepted: 01/09/2019] [Indexed: 12/15/2022]
Abstract
A novel bispecific α-Gal liposome was constructed by self-assembling AS1411 aptamers into the α-Gal containing liposomes. The α-Gal liposomes were prepared using cell membranes of red blood cells from rabbit, which are composed of cholesterol, phospholipids, and α-Gal glycolipids. AS1411 is a DNA aptamer with high specificity and affinity for nucleolin and could integrate into liposomes by the modification of cholesterol. The bispecific α-Gal liposomes surface-functionalized by α-Gal and AS1411 aptamer could recognize anti-Gal antibodies and nucleolin overexpressed by tumor cells simultaneously, followed by activating the immune system to attack the tumor cells, resulting in the lysis of the tumor cells by antibody dependent cell-mediated cytotoxicity. Under simulated tumor environment, the lysis rate of MCF-7 cells treated by the AS1411 modified α-Gal liposomes drastically increased compared to the liposomes without AS1411 aptamer. This study suggests that the AS1411 modified α-Gal liposomes can recognize nucleolin-overexpressing tumor cells selectively, subsequently improve the effect of the immunotherapy with high specificity. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1176-1183, 2019.
Collapse
Affiliation(s)
- Shanni Hong
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Pi Ding
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Yu Luo
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Tian Gao
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Ye Zhang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Renjun Pei
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| |
Collapse
|
27
|
Le DHT, Commandeur U, Steinmetz NF. Presentation and Delivery of Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand via Elongated Plant Viral Nanoparticle Enhances Antitumor Efficacy. ACS NANO 2019; 13:2501-2510. [PMID: 30668110 DOI: 10.1021/acsnano.8b09462] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Potato virus X (PVX) is a flexuous plant virus-based nanotechnology with promise in cancer therapy. As a high aspect ratio biologic (13 × 515 nm), PVX has excellent spatial control in structures and functions, offering high-precision nanoengineering for multivalent display of functional moieties. Herein, we demonstrate the preparation of the PVX-based nanocarrier for delivery of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a promising protein drug that induces apoptosis in cancer cells but not healthy cells. TRAIL bound to PVX by coordination bonds between nickel-coordinated nitrilotriacetic acid on PVX and His-tag on the protein could mimic the bioactive "membrane-bound" state in native TRAIL, resulting in an elongated nanoparticle displaying up 490 therapeutic protein molecules. Our data show that PVX-delivered TRAIL activates caspase-mediated apoptosis more efficiently compared to soluble TRAIL; also in vivo the therapeutic nanoparticle outperforms in delaying tumor growth in an athymic nude mouse model bearing human triple-negative breast cancer xenografts. This proof-of-concept work highlights the potential of filamentous plant virus nanotechnologies, particularly for targeting protein drug delivery for cancer therapy.
Collapse
Affiliation(s)
- Duc H T Le
- Department of Biomedical Engineering , Case Western Reserve University School of Medicine , Cleveland , Ohio 44106 , United States
| | - Ulrich Commandeur
- Department of Molecular Biology , RWTH-Aachen University , Aachen 52064 , Germany
| | - Nicole F Steinmetz
- Department of NanoEngineering, Moores Cancer Center, Department of Radiology, Department of Bioengineering , University of California, San Diego , La Jolla , California 92093 , United States
- Department of Biomedical Engineering , Case Western Reserve University School of Medicine , Cleveland , Ohio 44106 , United States
| |
Collapse
|
28
|
Pishavar E, Attaranzadeh A, Alibolandi M, Ramezani M, Hashemi M. Modified PAMAM vehicles for effective TRAIL gene delivery to colon adenocarcinoma: in vitro and in vivo evaluation. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S503-S513. [PMID: 30095012 DOI: 10.1080/21691401.2018.1500372] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
TRAIL (tumour necrosis factor-related apoptosis-inducing ligand) gene therapy is considered as one of the promising approaches for cancer treatment. Polyamidoamine (PAMAM) is one of the most extensively applied polymeric vector in gene delivery. In the current study, PAMAM (G4 and G5) dendrimers were modified with alkyl-carboxylate chain, PEG and cholesteryl chloroformate in order to enhance transfection efficiency through overcoming extracellular and intracellular barriers while reducing PAMAM cytotoxicity. Gene delivery efficiency of synthetized vectors was evaluated by both GFP (green fluorescent protein) reporter gene and TRAIL plasmid in colon cancer cells, in vitro and in vivo. The obtained results demonstrated that PAMAM G4-alkyl-PEG (3%)-Chol (5%)-TRAIL complexes at C/P ratio 4 could significantly increase cell death (29.45%) in comparison with unmodified PAMAM vector (15.5%). Moreover, in vivo study in C26 tumor-bearing BALB/c mice suggested that the prepared non-toxic safe vector could inhibit the tumor growth. This study represented the potent vehicle based on cholesterol-grafted PAMAM dendrimers with alkyl-PEG modification for efficient gene delivery in vitro and in vivo.
Collapse
Affiliation(s)
- Elham Pishavar
- a Pharmacutical Research Center, Pharmaceutical Technology Institute , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Armin Attaranzadeh
- b Department of Molecular Pathology and Cytogenetics , Mashhad University of Medical Sciences, Imam Reza Hospital , Mashhad , Iran
| | - Mona Alibolandi
- a Pharmacutical Research Center, Pharmaceutical Technology Institute , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Mohammad Ramezani
- a Pharmacutical Research Center, Pharmaceutical Technology Institute , Mashhad University of Medical Sciences , Mashhad , Iran.,c Department of Biotechnology, School of Pharmacy , Mashhad University of Medical Sciences , Mashhad , Iran
| | - Maryam Hashemi
- d Nanotechnology Research Center, Pharmaceutical Technology Institute , Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
29
|
Kih M, Lee EJ, Lee NK, Kim YK, Lee KE, Jeong C, Yang Y, Kim DH, Kim IS. Designed trimer-mimetic TNF superfamily ligands on self-assembling nanocages. Biomaterials 2018; 180:67-77. [PMID: 30025246 DOI: 10.1016/j.biomaterials.2018.07.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 12/19/2022]
Abstract
Presentation of an endogenous bioactive ligand in its native form is a key factor in controlling and determining its bioactivity, stability, and therapeutic efficacy. In this study, we developed a novel strategy for presenting trimeric ligands on nanocages by designing, optimizing and testing based on the rational design, high-resolution structural analysis and agonistic activity assays in vitro and in vivo. We successfully designed a nanocage that presents the TNF superfamily member, TRAIL (TNF-related apoptosis-inducing ligand) in its native-like trimeric structure. The native structure of TRAIL complexes was mimicked on the resulting trimeric TRAIL-presenting nanocages (TTPNs) by inserting sufficient spacing, determined from three-dimensional structural models, to provide optimal access to the corresponding receptors. The efficacy of TTPNs as an anti-tumor agent was confirmed in preclinical studies, which revealed up to 330-fold increased affinity, 62.5-fold enhanced apoptotic activity, and improved pharmacokinetic characteristics and stability compared with the monomeric form of TRAIL (mTRAIL). In this latter context, TTPNs exhibited greater than 90% stability over 1 mo, whereas ∼50% of mTRAIL aggregated within 2 d. Consistent with their enhanced stability and ultra-high affinity for the TRAIL receptor, TTPNs effectively induced apoptosis of tumor cells in vivo, leading to effective inhibition of tumor growth. Although TRAIL was used here as a proof-of-concept, all members of the TNF superfamily share the TNF homology domain (THD) and have similar distances between ecto-domain C-termini. Thus, other TNF superfamily ligands could be genetically substituted for the TRAIL ligand on the surface of this biomimetic delivery platform.
Collapse
Affiliation(s)
- Minwoo Kih
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Eun Jung Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Department of Chemical Engineering, School of Applied Chemical Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Na Kyeong Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Yoon Kyoung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Kyung Eun Lee
- Advanced Analysis Center, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Cherlhyun Jeong
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Yoosoo Yang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea.
| |
Collapse
|
30
|
Shi Y, Pang X, Wang J, Liu G. NanoTRAIL-Oncology: A Strategic Approach in Cancer Research and Therapy. Adv Healthc Mater 2018. [PMID: 29527836 DOI: 10.1002/adhm.201800053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
TRAIL is a member of the tumor necrosis factor superfamily that can largely trigger apoptosis in a wide variety of cancer cells, but not in normal cells. However, insufficient exposure to cancer tissues or cells and drug resistance has severely impeded the clinical application of TRAIL. Recently, nanobiotechnology has brought about a revolution in advanced drug delivery for enhanced anticancer therapy using TRAIL. With the help of materials science, immunology, genetic engineering, and protein engineering, substantial progress is made by expressing fusion proteins with TRAIL, engineering TRAIL on biological membranes, and loading TRAIL into functional nanocarriers or conjugating it onto their surfaces. Thus, the nanoparticle-based TRAIL (nanoTRAIL) opens up intriguing opportunities for efficient and safe bioapplications. In this review, the mechanisms of action and biological function of TRAIL, as well as the current status of TRAIL treatment, are comprehensively discussed. The application of functional nanotechnology combined with TRAIL in cancer therapy is also discussed.
Collapse
Affiliation(s)
- Yesi Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine; School of Public Health; Xiamen University; Xiamen 361102 China
| | - Xin Pang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine; School of Public Health; Xiamen University; Xiamen 361102 China
| | - Junqing Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine; School of Public Health; Xiamen University; Xiamen 361102 China
- Collaborative Innovation Center of Guangxi Biological Medicine and the; Medical and Scientific Research Center; Guangxi Medical University; Nanning 530021 China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine; School of Public Health; Xiamen University; Xiamen 361102 China
| |
Collapse
|
31
|
Pro-apoptotic peptides-based cancer therapies: challenges and strategies to enhance therapeutic efficacy. Arch Pharm Res 2018; 41:594-616. [PMID: 29804279 DOI: 10.1007/s12272-018-1038-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 05/10/2018] [Indexed: 12/30/2022]
Abstract
Cancer is a leading cause of death worldwide. Despite many advances in the field of cancer therapy, an effective cure is yet to be found. As a more potent alternative for the conventional small molecule anti-cancer drugs, pro-apoptotic peptides have emerged as a new class of anticancer agents. By interaction with certain members in the apoptotic pathways, they could effectively kill tumor cells. However, there remain bottleneck challenges for clinical application of these pro-apoptotic peptides in cancer therapy. In this review, we will overview the developed pro-apoptotic peptides and outline the widely adopted molecular-based and nanoparticle-based strategies to enhance their anti-tumor effects.
Collapse
|
32
|
Sun T, Zhu T, Liang X, Yang S, Zhao R. Effects of Recombinant Circularly Permuted Tumor Necrosis Factor (TNF)-Related Apoptosis-Inducing Ligand (TRAIL) (Recombinant Mutant Human TRAIL) in Combination with 5-Fluorouracil in Human Colorectal Cancer Cell Lines HCT116 and SW480. Med Sci Monit 2018; 24:2550-2561. [PMID: 29695684 PMCID: PMC5939707 DOI: 10.12659/msm.909390] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background Circularly permuted tumor necrosis factor-related apoptosis-inducing ligand, a mutant form of tumor necrosis factor-related apoptosis-inducing ligand, is an effective antitumor cytokine. However, its antitumor effect in colorectal cancer is unclear. This study assessed the antitumor effect of circularly permuted tumor necrosis factor-related apoptosis-inducing ligand alone or with 5-fluorouracil in colorectal cancer cells in vitro and explored the underlying mechanisms. Material/Methods We used the (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (MTS) assay to analyze cell proliferation inhibition. The apoptotic effects of circularly permuted tumor necrosis factor-related apoptosis-inducing ligand, 5-fluorouracil, or both in human colorectal cancer cells were evaluated using flow cytometry. Furthermore, the levels of apoptosis-related proteins were examined by Western blotting. Results Compared to either agent alone, cotreatment with 5-fluorouracil and circularly permuted tumor necrosis factor-related apoptosis-inducing ligand showed obvious antitumor effects and induced significant apoptosis of colorectal cancer cells. 5-Fluorouracil enhanced circularly permuted tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis by increasing death receptor 4 and 5 levels in HCT116 cells, but only of death receptor 4 in SW480 cells. Moreover, 5-fluorouracil plus circularly permuted tumor necrosis factor-related apoptosis-inducing ligand increased apoptosis-related protein levels such as cleaved caspase-3, caspase-8, and poly-ADP-ribose polymerase and downregulated that of the survival protein B-cell lymphoma-extra-large. Pretreatment with the pan-caspase inhibitor, z-VAD-FMK, attenuated the caspase-dependent apoptosis induced by circularly permuted tumor necrosis factor-related apoptosis-inducing ligand alone or combined with 5-fluorouracil. Conclusions Cotreatment with 5-fluorouracil and circularly permuted tumor necrosis factor-related apoptosis-inducing ligand showed enhanced antitumor effects on colorectal cancer cells.
Collapse
Affiliation(s)
- Tongyou Sun
- Department of Oncology, Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| | - Tienian Zhu
- Key Laboratory of Immune Mechanism and Intervention of Serious Diseases in Hebei Province, Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, China (mainland).,Department of Medical Oncology, Bethune International Peace Hospital, Shijiazhuang, Hebei, China (mainland)
| | - Xiujun Liang
- Basic Medical Institute, Chengde Medical University, Chengde, Hebei, China (mainland)
| | - Shifang Yang
- Beijing Sunbio Biotech Co., Ltd., Beijing, China (mainland)
| | - Ruijing Zhao
- Key Laboratory of Immune Mechanism and Intervention of Serious Diseases in Hebei Province, Department of Immunology, Hebei Medical University, Shijiazhuang, Hebei, China (mainland)
| |
Collapse
|
33
|
Dubuisson A, Micheau O. Antibodies and Derivatives Targeting DR4 and DR5 for Cancer Therapy. Antibodies (Basel) 2017; 6:E16. [PMID: 31548531 PMCID: PMC6698863 DOI: 10.3390/antib6040016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/16/2017] [Accepted: 10/19/2017] [Indexed: 02/07/2023] Open
Abstract
Developing therapeutics that induce apoptosis in cancer cells has become an increasingly attractive approach for the past 30 years. The discovery of tumor necrosis factor (TNF) superfamily members and more specifically TNF-related apoptosis-inducing ligand (TRAIL), the only cytokine of the family capable of eradicating selectively cancer cells, led to the development of numerous TRAIL derivatives targeting death receptor 4 (DR4) and death receptor 5 (DR5) for cancer therapy. With a few exceptions, preliminary attempts to use recombinant TRAIL, agonistic antibodies, or derivatives to target TRAIL agonist receptors in the clinic have been fairly disappointing. Nonetheless, a tremendous effort, worldwide, is being put into the development of novel strategic options to target TRAIL receptors. Antibodies and derivatives allow for the design of novel and efficient agonists. We summarize and discuss here the advantages and drawbacks of the soar of TRAIL therapeutics, from the first developments to the next generation of agonistic products, with a particular insight on new concepts.
Collapse
Affiliation(s)
- Agathe Dubuisson
- University Bourgogne Franche-Comté, INSERM, LNC UMR1231, F-21079 Dijon, France.
- CovalAb, Research Department, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France.
- INSERM, UMR1231, Laboratoire d'Excellence LipSTIC, F-21079 Dijon, France.
| | - Olivier Micheau
- University Bourgogne Franche-Comté, INSERM, LNC UMR1231, F-21079 Dijon, France.
- CovalAb, Research Department, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France.
- INSERM, UMR1231, Laboratoire d'Excellence LipSTIC, F-21079 Dijon, France.
| |
Collapse
|
34
|
Wu X, Wang S, Li M, Wang A, Zhou Y, Li P, Wang Y. Nanocarriers for TRAIL delivery: driving TRAIL back on track for cancer therapy. NANOSCALE 2017; 9:13879-13904. [PMID: 28914952 DOI: 10.1039/c7nr04959e] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Since its initial identification, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has been shown to be capable of selectively inducing apoptosis in cancer cells. However, translation of the encouraging preclinical studies of this cytokine into the clinic has been restricted by its extremely short half-life, the presence of resistant cancer cell populations, and its inefficient in vivo delivery. Recently, there has been exceptional progress in developing novel formulations to increase the circulatory half-life of TRAIL and new combinations to treat cancers that are resistant to TRAIL. In particular, TRAIL-based nanotherapies offer the potential to improve the stability of TRAIL and prolong its half-life in plasma, to specifically deliver TRAIL to a particular target site, and to overcome resistance to TRAIL. The aim of this review is to provide an overview of the state-of-the art drug delivery systems that are currently being tested or developed to improve the biological attributes of TRAIL-based therapies.
Collapse
Affiliation(s)
- Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan Province, China
| | | | | | | | | | | | | |
Collapse
|