1
|
Wu D, Li Y, Dai Y, Tian H, Chen Y, Shen G, Yang G. Stabilization of chitosan-based nanomedicines in cancer therapy: a review. Int J Biol Macromol 2025; 309:143016. [PMID: 40216118 DOI: 10.1016/j.ijbiomac.2025.143016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/27/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Chitosan (CS), a versatile and alkaline polysaccharide, has gained significant attention in nanomedicine due to its biocompatibility and biodegradability. In recent years, its applications in cancer therapy, particularly for the delivery of chemotherapeutic drugs, diagnostic agents, and genes, have advanced considerably. However, many CS-based nanomedicines suffer from poor stability in biological fluids, especially under physiological conditions. The neutral pH and the presence of electrolytes in physiological environments reduce the charge density of CS, which can account for this application limitation of CS-based nanomedicines. To improve the stability and prevent dissociation or aggregation of these nanomedicines before reaching the target sites, this review summarizes common stabilization strategies including hydrophilic or hydrophobic modification of CS, as well as incorporation with metal ions (e.g. Fe3+ or Zn2+), complexation with anionic cross-linkers (e.g. TPP) or anionic polymers. Additionally, the review highlights the application of stabilized CS-based nanocarriers in drug delivery, with a particular focus on cancer therapy. The challenges and future perspectives for accelerating the clinical translation of these nanomedicines are also discussed.
Collapse
Affiliation(s)
- Danjun Wu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yazhen Li
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yiwei Dai
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hong Tian
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yifei Chen
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Gongmin Shen
- Hangzhou Guoguang Pharmaceutical Co., Ltd., Hangzhou 310018, China.
| | - Gensheng Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
2
|
Alemi PS, Mohamadali M, Arabahmadi S, Irani S, Sharifi F. Carboxymethyl Chitosan and Chitosan as a Bioactive Delivery System: A Review. Biotechnol Appl Biochem 2025:e2758. [PMID: 40275440 DOI: 10.1002/bab.2758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/16/2025] [Indexed: 04/26/2025]
Abstract
The functionality and mechanism of bioactive agents (BA) in treating various diseases have been studied as a progressive route. Designing an effective delivery system for transferring these molecules and components is a major challenge. For that reason, a wide range of biomaterials has been introduced to deliver BA to the target tissue or cells. Chitosan (CTS) is a nontoxic, biocompatible, biodegradable, and notable point low-cost polymer, and, as a result, can be effectively utilized in the formulation of diverse delivery systems, in biomedical applications. However, CTS has some limitations, such as poor solubility in aqueous and alkaline media, rapid swelling and degradation, and consequence fast release agent. The CTS derivative carboxymethyl chitosan (CMC) is an acceptable candidate for overcoming these limitations. CMC is a high-impact grade for pharmaceutical and biomedical applications because of its nontoxic, biocompatible, biodegradable, gelation, mucoadhesive, antibacterial, and antifungal. CMC bioactivity potentials are related to carboxyl and methyl groups added through chemical modification in the CTS backbone. In this review, the physical and chemical properties of CTS and CMC have been introduced and discussed. Afterward, its biomedical applications with delivery approaches for various BA (drugs, genes, proteins), microfluidic, and cancer have been considered.
Collapse
Affiliation(s)
- Parinaz Sadat Alemi
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Marjan Mohamadali
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Samira Arabahmadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Fereshteh Sharifi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
- Soft Tissue Engineering Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
3
|
Vetter VC, Yazdi M, Gialdini I, Pöhmerer J, Seidl J, Höhn M, Lamb DC, Wagner E. Ionic Coating of siRNA Polyplexes with cRGD-PEG-Hyaluronic Acid To Modulate Serum Stability and In Vivo Performance. Biochemistry 2025; 64:1509-1529. [PMID: 40102188 DOI: 10.1021/acs.biochem.4c00650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Efficient delivery of siRNA-based polyplexes to tumors remains a major challenge. Nonspecific interactions in the bloodstream, limited circulation time, and nontargeted biodistribution hamper sufficient tumor accumulation. To address these challenges, we developed an ionic hyaluronic acid (HA) coating to shield sequence-defined oligoaminoamide-based polyplexes. This coating should shield the positive polyplex surface charge, thus reducing nonspecific interactions and enhancing serum stability. Additionally, we modified the HA coating with the cyclic RGDfK (cRGD) peptide to specifically target tumor endothelial cells (TECs). Optionally, a polyethylene glycol (PEG) spacer was also introduced to improve ligand presentation on the polyplex surface. The HA-coated polyplexes exhibited favorable physicochemical properties, including a negative zeta potential and effective siRNA retention within the polyplex, which was not adversely affected by PEG or cRGD modification. In vitro analyses revealed that these polyplexes not only enhanced tumor cell association and preserved the high transfection efficiency of plain cationic polyplexes but also exhibited coating-dependent cellular internalization, as evidenced by a competitive inhibition experiment. Even in the presence of serum, the HA-coated polyplexes encapsulated siRNA effectively, exhibited suitable particle sizes, and maintained a high gene silencing efficiency. In vivo studies involving intravenous administration into Neuro2a tumor-bearing mice showed that the HA coating, particularly when modified with PEG and cRGD, significantly increased the tumor accumulation of polyplexes. HA-PEG-cRGD-shielded polyplexes exhibited significantly enhanced in vivo gene silencing in tumors compared with plain polyplexes. Collectively, our results indicate a superior performance of HA-coated polyplexes in terms of stability and cellular uptake, both in vitro and in vivo.
Collapse
Affiliation(s)
- Victoria C Vetter
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
| | - Mina Yazdi
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
| | - Irene Gialdini
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, Munich 81377, Germany
| | - Jana Pöhmerer
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
| | - Johanna Seidl
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
| | - Don C Lamb
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, Munich 81377, Germany
- Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
- Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
- CNATM─Cluster for Nucleic Acid Therapeutics, Würmtalstr. 201, Munich 81377, Germany
| |
Collapse
|
4
|
Zhang Z, Tang Y, Luo D, Qiu J, Chen L. Advances in nanotechnology for targeting cancer-associated fibroblasts: A review of multi-strategy drug delivery and preclinical insights. APL Bioeng 2025; 9:011502. [PMID: 40094065 PMCID: PMC11910205 DOI: 10.1063/5.0244706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs) play a crucial role in the tumor microenvironment by promoting tumor growth, immune evasion, and metastasis. Recently, drug delivery systems targeting CAFs have emerged as a promising long-term and effective approach to cancer treatment. Advances in nanotechnology, in particular, have led to the development of nanomedicine delivery systems designed specifically to target CAFs, offering new possibilities for precise and personalized cancer therapies. This article reviews recent progress in drug delivery using nanocarriers that target CAFs. Additionally, we explore the potential of combining multiple therapies, such as chemotherapy and immunotherapy, with nanocarriers to enhance efficacy and overcome drug resistance. Although many preclinical studies show promise, the clinical application of nanomedicine still faces considerable challenges, especially in terms of drug penetration and large-scale production. Therefore, this review aims to provide a fresh perspective on CAF-targeted drug delivery systems and highlight potential future research directions and clinical applications.
Collapse
|
5
|
Umar AK, Limpikirati PK, Rivai B, Ardiansah I, Sriwidodo S, Luckanagul JA. Complexed hyaluronic acid-based nanoparticles in cancer therapy and diagnosis: Research trends by natural language processing. Heliyon 2025; 11:e41246. [PMID: 39811313 PMCID: PMC11729671 DOI: 10.1016/j.heliyon.2024.e41246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Hyaluronic acid (HA) is a popular surface modifier in targeted cancer delivery due to its receptor-binding abilities. However, HA alone faces limitations in lipid solubility, biocompatibility, and cell internalization, making it less effective as a standalone delivery system. This comprehensive study aimed to explore a dynamic landscape of complexation in HA-based nanoparticles in cancer therapy, examining diverse aspects from influential modifiers to emerging trends in cancer diagnostics. We discovered that certain active substances, such as 5-aminolevulinic acid, adamantane, and protamine, have been on trend in terms of their usage over the past decade. Dextran, streptavidin, and catechol emerge as intriguing conjugates for HA, coupled with nanostar, quantum dots, and nanoprobe structures for optimal drug delivery and diagnostics. Strategies like hypoxic conditioning, dual responsiveness, and pulse laser activation enhance controlled release, targeted delivery, and real-time diagnostic techniques like ultrasound imaging and X-ray computed tomography (X-ray CT). Based on our findings, conventional bibliometric tools fail to highlight relevant topics in this area, instead producing merely abstract and broad-meaning keywords. Extraction using Named Entity Recognition and topic search with Latent Dirichlet Allocation successfully revealed five representative topics with the ability to exclude irrelevant keywords. A shift in research focuses from optimizing chemical toxicity to particular targeting tactics and precise release mechanisms is evident. These findings reflect the dynamic landscape of HA-based nanoparticle research in cancer therapy, emphasizing advancements in targeted drug delivery, therapeutic efficacy, and multimodal diagnostic approaches to improve overall patient outcomes.
Collapse
Affiliation(s)
- Abd Kakhar Umar
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Medical Informatics Laboratory, ETFLIN, Palu City, 94225, Indonesia
| | - Patanachai K. Limpikirati
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Metabolomics for Life Sciences Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Bachtiar Rivai
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Medical Informatics Laboratory, ETFLIN, Palu City, 94225, Indonesia
| | - Ilham Ardiansah
- Department of Animal Husbandry, Faculty Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Medical Informatics Laboratory, ETFLIN, Palu City, 94225, Indonesia
| | - Sriwidodo Sriwidodo
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Jittima Amie Luckanagul
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
6
|
Bian X, Yu X, Lu S, Jia L, Li P, Yin J, Tan S. Chitosan-based nanoarchitectures for siRNA delivery in cancer therapy: A review of pre-clinical and clinical importance. Int J Biol Macromol 2025; 284:137708. [PMID: 39571854 DOI: 10.1016/j.ijbiomac.2024.137708] [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: 10/12/2024] [Revised: 11/01/2024] [Accepted: 11/13/2024] [Indexed: 11/30/2024]
Abstract
The gene therapy has been developed into a new cancer treatment option. Now that we know which molecular components contribute to carcinogenesis, we may use gene therapy to target particular signalling pathways in cancer treatment. Problems with gene therapy include genetic tool degradation in blood, off-targeting features, and inadequate tumor site accumulation; new delivery mechanisms are needed to address these issues. A polysaccharide made from chitin, chitosan has found extensive use in the creation of nanoparticles. The delivery of genes in the treatment of illnesses, particularly cancer, has made use of nanostructures modified with chitosan. Topics covered in this review center on cancer treatment using chitosan-based polymers for siRNA delivery. This study aims to assess the potential of chitosan nanoparticles for the simultaneous administration of siRNA and anti-cancer medications. In cancer treatment, these nanoparticles can transport phytochemicals or chemotherapeutics together with siRNA. In addition, chitosan nanoparticles loaded with siRNA can inhibit the growth and spread of human malignancies by delivering siRNA that targets particular genes. Chitosan nanoparticles loaded with siRNA can heighten the responsiveness of cancer cells. Future therapeutic applications of chitosan nanoparticles may open the path for cancer treatment, thanks to their biocompatibility and biosafety.
Collapse
Affiliation(s)
- Xiaobo Bian
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaopeng Yu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shiyang Lu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Linan Jia
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ping Li
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Jianqiao Yin
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Shutao Tan
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
7
|
Liu J, Xi Z, Fan C, Mei Y, Zhao J, Jiang Y, Zhao M, Xu L. Hydrogels for Nucleic Acid Drugs Delivery. Adv Healthc Mater 2024; 13:e2401895. [PMID: 39152918 DOI: 10.1002/adhm.202401895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/05/2024] [Indexed: 08/19/2024]
Abstract
Nucleic acid drugs are one of the hot spots in the field of biomedicine in recent years, and play a crucial role in the treatment of many diseases. However, its low stability and difficulty in target drug delivery are the bottlenecks restricting its application. Hydrogels are proven to be promising for improving the stability of nucleic acid drugs, reducing the adverse effects of rapid degradation, sudden release, and unnecessary diffusion of nucleic acid drugs. In this review, the strategies of loading nucleic acid drugs in hydrogels are summarized for various biomedical research, and classify the mechanism principles of these strategies, including electrostatic binding, hydrogen bond based binding, hydrophobic binding, covalent bond based binding and indirect binding using various carriers. In addition, this review also describes the release strategies of nucleic acid drugs, including photostimulation-based release, enzyme-responsive release, pH-responsive release, and temperature-responsive release. Finally, the applications and future research directions of hydrogels for delivering nucleic acid drugs in the field of medicine are discussed.
Collapse
Affiliation(s)
- Jiaping Liu
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Ziyue Xi
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Chuanyong Fan
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Yihua Mei
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Jiale Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Yingying Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Ming Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Lu Xu
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| |
Collapse
|
8
|
Naghib SM, Ahmadi B, Mikaeeli Kangarshahi B, Mozafari MR. Chitosan-based smart stimuli-responsive nanoparticles for gene delivery and gene therapy: Recent progresses on cancer therapy. Int J Biol Macromol 2024; 278:134542. [PMID: 39137858 DOI: 10.1016/j.ijbiomac.2024.134542] [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: 04/17/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 08/15/2024]
Abstract
Recent cancer therapy research has found that chitosan (Ch)-based nanoparticles show great potential for targeted gene delivery. Chitosan, a biocompatible and biodegradable polymer, has exceptional properties, making it an ideal carrier for therapeutic genes. These nanoparticles can respond to specific stimuli like pH, temperature, and enzymes, enabling precise delivery and regulated release of genes. In cancer therapy, these nanoparticles have proven effective in delivering genes to tumor cells, slowing tumor growth. Adjusting the nanoparticle's surface, encapsulating protective agents, and using targeting ligands have also improved gene delivery efficiency. Smart nanoparticles based on chitosan have shown promise in improving outcomes by selectively releasing genes in response to tumor conditions, enhancing targeted delivery, and reducing off-target effects. Additionally, targeting ligands on the nanoparticles' surface increases uptake and effectiveness. Although further investigation is needed to optimize the structure and composition of these nanoparticles and assess their long-term safety, these advancements pave the way for innovative gene-focused cancer therapies.
Collapse
Affiliation(s)
- Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran 1684613114, Iran.
| | - Bahar Ahmadi
- Biomaterials and Tissue Engineering Research Group, Interdisciplinary Technologies Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Babak Mikaeeli Kangarshahi
- State Key Laboratory of Structure Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
9
|
Antoniou V, Mourelatou EA, Galatou E, Avgoustakis K, Hatziantoniou S. Gene Therapy with Chitosan Nanoparticles: Modern Formulation Strategies for Enhancing Cancer Cell Transfection. Pharmaceutics 2024; 16:868. [PMID: 39065565 PMCID: PMC11280172 DOI: 10.3390/pharmaceutics16070868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Gene therapy involves the introduction of exogenous genetic material into host tissues to modify gene expression or cellular properties for therapeutic purposes. Initially developed to address genetic disorders, gene therapy has expanded to encompass a wide range of conditions, notably cancer. Effective delivery of nucleic acids into target cells relies on carriers, with non-viral systems gaining prominence due to their enhanced safety profile compared to viral vectors. Chitosan, a biopolymer, is frequently utilized to fabricate nanoparticles for various biomedical applications, particularly nucleic acid delivery, with recent emphasis on targeting cancer cells. Chitosan's positively charged amino groups enable the formation of stable nanocomplexes with nucleic acids and facilitate interaction with cell membranes, thereby promoting cellular uptake. Despite these advantages, chitosan-based nanoparticles face challenges such as poor solubility at physiological pH, non-specificity for cancer cells, and inefficient endosomal escape, limiting their transfection efficiency. To address these limitations, researchers have focused on enhancing the functionality of chitosan nanoparticles. Strategies include improving stability, enhancing targeting specificity, increasing cellular uptake efficiency, and promoting endosomal escape. This review critically evaluates recent formulation approaches within these categories, aiming to provide insights into advancing chitosan-based gene delivery systems for improved efficacy, particularly in cancer therapy.
Collapse
Affiliation(s)
- Varvara Antoniou
- Pharmacy Program, Department of Health Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus; (V.A.); (E.G.)
| | - Elena A. Mourelatou
- Pharmacy Program, Department of Health Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus; (V.A.); (E.G.)
- Bioactive Molecules Research Center, School of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus
| | - Eleftheria Galatou
- Pharmacy Program, Department of Health Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus; (V.A.); (E.G.)
- Bioactive Molecules Research Center, School of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus
| | - Konstantinos Avgoustakis
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, University of Patras, 26 504 Rion, Greece; (K.A.); (S.H.)
| | - Sophia Hatziantoniou
- Laboratory of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, University of Patras, 26 504 Rion, Greece; (K.A.); (S.H.)
| |
Collapse
|
10
|
Sohrabi S, Alipour S, Ghahramanipour Z, Masoumi J, Baradaran B. STAT signaling pathways in immune cells and their associated mechanisms in cancer pathogenesis. BIOIMPACTS : BI 2024; 15:30030. [PMID: 39963570 PMCID: PMC11830145 DOI: 10.34172/bi.30030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/11/2023] [Accepted: 10/28/2023] [Indexed: 02/20/2025]
Abstract
Introduction Signal transducer and activator of transcriptions (STATs) factors as critical proteins in cell signaling regulate diverse biological processes such as differentiation and proliferation of cells. STATs have been shown to play distinct roles in modulating immune responses mediated by innate and adaptive immune cell subsets due to their significant roles in cytokine signaling. Methods In the current study, we review recent studies on the contribution of individual STAT proteins to cytokine signaling, development, and activity of diverse immune cells that constitute the whole immune system and help its performance against endogenous or exogenous agents with a particular focus on meaningful STAT factor in each of innate and adaptive immune cells' subsets to clarify their function in favor of the tumor or against it. Results Dysregulation of signaling pathways in the immune cells is associated with various immune disorders, such as the inability of immune system cells in the effective destruction of cancerous cells. Increase of knowledge about these pathways' functions is essential to understand how they can be effectively targeted to eliminate tumors. Conclusion The majority of immune cells use the Jak/STAT signaling pathway, which is one of the most important signaling pathways with a role in induction of proper immune responses. Since each of the STAT factors has a specific role in diverse immune cells' subsets, appropriate targeting of them can be a promising strategy for patients who suffer from immune system disorders; specifically it can be beneficial as an approach for cancer immunotherapy.
Collapse
Affiliation(s)
- Sepideh Sohrabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Alipour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Tabasi H, Mollazadeh S, Fazeli E, Abnus K, Taghdisi SM, Ramezani M, Alibolandi M. Transitional Insight into the RNA-Based Oligonucleotides in Cancer Treatment. Appl Biochem Biotechnol 2024; 196:1685-1711. [PMID: 37402038 DOI: 10.1007/s12010-023-04597-5] [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] [Accepted: 06/19/2023] [Indexed: 07/05/2023]
Abstract
Conventional cancer therapies with chemodrugs suffer from various disadvantages, such as irreversible side effects on the skin, heart, liver, and nerves with even fatal consequences. RNA-based therapeutic is a novel technology which offers great potential as non-toxic, non-infectious, and well-tolerable platform. Herein, we introduce different RNA-based platforms with a special focus on siRNA, miRNA, and mRNA applications in cancer treatment in order to better understand the details of their therapeutic effects. Of note, the co-delivery of RNAs with other distinct RNA or drugs has provided safe, efficient, and novel treatment modalities for cancer treatment.
Collapse
Affiliation(s)
- Hamed Tabasi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Elham Fazeli
- Biomedicine Department, Aarhus University, Aarhus, Denmark
| | - Khalil Abnus
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Gherman A, Bolundut D, Ecea R, Balacescu L, Curcean S, Dina C, Balacescu O, Cainap C. Molecular Subtypes, microRNAs and Immunotherapy Response in Metastatic Colorectal Cancer. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:397. [PMID: 38541123 PMCID: PMC10972200 DOI: 10.3390/medicina60030397] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/10/2024] [Accepted: 02/18/2024] [Indexed: 11/12/2024]
Abstract
Currently, only a limited set of molecular traits are utilized to direct treatment for metastatic CRC (mCRC). The molecular classification of CRC depicts tumor heterogeneity based on gene expression patterns and aids in comprehending the biological characteristics of tumor formation, growth and prognosis. Additionally, it assists physicians in tailoring the therapeutic approach. Microsatellite instability (MSI-H)/deficient mismatch repair proteins (MMRd) status has become a ubiquitous biomarker in solid tumors, caused by mutations or methylation of genes and, in turn, the accumulation of mutations and antigens that subsequently induce an immune response. Immune checkpoint inhibitors (ICI) have recently received approval for the treatment of mCRC with MSI-H/MMRd status. However, certain individuals experience either initial or acquired resistance. The tumor-programmed cell death ligand 1 (PD-L1) has been linked to the ability of CRC to evade the immune system and promote its growth. Through comprehensive research conducted via the PUBMED database, the objectives of this paper were to review the molecular characteristics linked to tumor response in metastatic CRC in light of improved patients' outcomes following ICI therapies as seen in clinical trials and to identify particular microRNAs that can modulate the expression of specific oncoproteins, such as PD-L1, and disrupt the mechanisms that allow the immune system to be evaded.
Collapse
Affiliation(s)
- Alexandra Gherman
- 10th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (A.G.); (C.C.)
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (D.B.); (R.E.)
| | - Dinu Bolundut
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (D.B.); (R.E.)
| | - Radu Ecea
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (D.B.); (R.E.)
| | - Loredana Balacescu
- Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania;
| | - Sebastian Curcean
- 10th Department of Radiation Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania;
- Department of Radiation Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Constantin Dina
- Department of Anatomy, Faculty of Medicine, Ovidius University, 124 Mamaia Boulevard, 900527 Constanta, Romania
| | - Ovidiu Balacescu
- Department of Genetics, Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania;
| | - Calin Cainap
- 10th Department of Medical Oncology, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (A.G.); (C.C.)
- Department of Medical Oncology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (D.B.); (R.E.)
| |
Collapse
|
13
|
Zaiki Y, Iskandar A, Wong TW. Functionalized chitosan for cancer nano drug delivery. Biotechnol Adv 2023; 67:108200. [PMID: 37331671 DOI: 10.1016/j.biotechadv.2023.108200] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/18/2023] [Accepted: 06/11/2023] [Indexed: 06/20/2023]
Abstract
Chitosan is a biotechnological derivative of chitin receiving a widespread pharmaceutical and biomedical applications. It can be used to encapsulate and deliver cancer therapeutics with inherent pH-dependent solubility to confer drug targeting at tumour microenvironment and anti-cancer activity synergizing cancer cytotoxic drug actions. To further reduce the off-target and by-stander adverse effects of drugs, a high targeted drug delivery efficiency at the lowest possible drug doses is clinically required. The chitosan has been functionalized with covalent conjugates or complexes and processed into nanoparticles to encapsulate and control drug release, to avoid premature drug clearance, to deliver drugs passively and actively to cancer site at tissue, cell or subcellular levels, and to promote cancer cell uptake of nanoparticles through membrane permeabilization at higher specificity and scale. Nanomedicine developed using functionalized chitosan translates to significant preclinical improvements. Future challenges related to nanotoxicity, manufacturability, selection precision of conjugates and complexes as a function of cancer omics and their biological responses from administration site to cancer target need critical assessments.
Collapse
Affiliation(s)
- Yazid Zaiki
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia; Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia
| | - Athirah Iskandar
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia; Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia
| | - Tin Wui Wong
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia; Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia; Sino-Malaysia Molecular Oncology and Traditional Chinese Medicine Delivery Joint Research Centre, Medical College, Yangzhou University, 136, Jiangyang Middle Road, Yangzhou, Jiangsu Province, China; Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
14
|
Tian B, Hua S, Liu J. Multi-functional chitosan-based nanoparticles for drug delivery: Recent advanced insight into cancer therapy. Carbohydr Polym 2023; 315:120972. [PMID: 37230614 DOI: 10.1016/j.carbpol.2023.120972] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/26/2023] [Accepted: 04/29/2023] [Indexed: 05/27/2023]
Abstract
Cancer therapy continues to be a major global concern, with conventional treatments suffering from low efficacy, untargeted drug delivery, and severe side effects. Recent research in nanomedicine suggests that nanoparticles' unique physicochemical properties can be leveraged to surmount the limitations of conventional cancer treatment. Chitosan-based nanoparticles have gained significant attention due to their high drug-carrying capacity, non-toxicity, biocompatibility, and long circulation time. Chitosan is utilized in cancer therapies as a carrier to accurately deliver active ingredients to tumor sites. This review focuses on clinical studies and current market offerings of anticancer drugs. The unique nature of tumor microenvironments presents new opportunities for the development of smart drug delivery systems, and this review explores the design and preparation of chitosan-based smart nanoparticles. Further, we discuss the therapeutic efficacies of these nanoparticles based on various in vitro and in vivo findings. Finally, we present a forward-looking perspective on the challenges and prospects of chitosan-based nanoparticles in cancer therapy, intending to provide fresh ideas for advancing cancer treatment strategies.
Collapse
Affiliation(s)
- Bingren Tian
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China; Key Laboratory of Ningxia Stem Cell and Regenerative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Shiyao Hua
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| | - Jiayue Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao.
| |
Collapse
|
15
|
Khodakarami A, Kashani MA, Nazer A, Kheshti AM, Rashidi B, Karpisheh V, Masjedi A, Abolhasani S, Izadi S, Bagherifar R, Hejazian SS, Mohammadi H, Movassaghpour A, Feizi AAH, Hojjat-Farsangi M, Jadidi-Niaragh F. Targeted Silencing of NRF2 by rituximab-conjugated nanoparticles increases the sensitivity of chronic lymphoblastic leukemia cells to Cyclophosphamide. Cell Commun Signal 2023; 21:188. [PMID: 37528446 PMCID: PMC10391779 DOI: 10.1186/s12964-023-01213-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 07/01/2023] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND Targeting influential factors in resistance to chemotherapy is one way to increase the effectiveness of chemotherapeutics. The nuclear factor erythroid 2-related factor 2 (Nrf2) pathway overexpresses in chronic lymphocytic leukemia (CLL) cells and appears to have a significant part in their survival and chemotherapy resistance. Here we produced novel nanoparticles (NPs) specific for CD20-expressing CLL cells with simultaneous anti-Nrf2 and cytotoxic properties. METHODS Chitosan lactate (CL) was used to produce the primary NPs which were then respectively loaded with rituximab (RTX), anti-Nrf2 Small interfering RNA (siRNAs) and Cyclophosphamide (CP) to prepare the final version of the NPs (NP-Nrf2_siRNA-CP). All interventions were done on both peripheral blood mononuclear cells (PBMCs) and bone marrow mononuclear cells (BMNCs). RESULTS NP-Nrf2_siRNA-CP had satisfying physicochemical properties, showed controlled anti-Nrf2 siRNA/CP release, and were efficiently transfected into CLL primary cells (both PBMCs and BMNCs). NP-Nrf2_siRNA-CP were significantly capable of cell apoptosis induction and proliferation prevention marked by respectively decreased and increased anti-apoptotic and pro-apoptotic factors. Furthermore, use of anti-Nrf2 siRNA was corresponding to elevated sensitivity of CLL cells to CP. CONCLUSION Our findings imply that the combination therapy of malignant CLL cells with RTX, CP and anti-Nrf2 siRNA is a novel and efficient therapeutic strategy that was capable of destroying malignant cells. Furthermore, the use of NPs as a multiple drug delivery method showed fulfilling properties; however, the need for further future studies is undeniable. Video Abstract.
Collapse
Affiliation(s)
- Atefeh Khodakarami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Atefeh Nazer
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Bentolhoda Rashidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Karpisheh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Masjedi
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Shiva Abolhasani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Izadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rafieh Bagherifar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - AliAkbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Zhang J, Jiang S, Li S, Jiang J, Mei J, Chen Y, Ma Y, Liu Y, Liu Y. Nanotechnology: A New Strategy for Lung Cancer Treatment Targeting Pro-Tumor Neutrophils. ENGINEERING 2023; 27:106-126. [DOI: 10.1016/j.eng.2022.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
17
|
Maimaiti A, Aierken Y, Zhou L, He J, Abudureyimu A, Li SX. Inhibiting Interleukin-6/Signal Transducers and Activators of Transduction-3/Hypoxia-Inducible Factor-1α Signaling Pathway Suppressed the Growth of Infantile Hemangioma. Eur J Pediatr Surg 2023; 33:158-166. [PMID: 35820438 DOI: 10.1055/s-0042-1749436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE This study aims to evaluate the expression of interleukin 6 (IL-6) in patients with infantile hemangioma (IH) and investigate the role of the IL-6/signal transducers and activators of transduction-3 (STAT3)/hypoxia-inducible factor-1α (HIF-1α) pathways in the progression of IH. METHODS Serum samples were obtained from the patients with IH and normal infants to measure IL-6 expression. Hemangioma-derived stem cells (HemSCs) were transfected with small interfering RNA (siRNA) targeting IL-6, HIF-1α, or STAT3. Then, cell viability and wound healing assays were conducted. After that, the HemSC tumor mouse model was established. The in vivo anticancer effect of the IL-6 inhibitor was investigated. RESULTS The patients with IH had much higher IL-6 levels compared with the healthy controls (p = 0.005). HemSCs transfected with IL-6 siRNA had significantly lower viability and migration rates than normal HemSCs. HemSCs transfected with STAT3 siRNA or HIF-1α siRNA had similar tendencies. On tumor-bearing mice, the IL-6 inhibitor treatment significantly delayed tumor growth. Compared with the control group, caspase-3 was significantly increased in the IL-6 inhibitor group (p < 0.05), whereas Ki-67 was decreased in the IL-6 inhibitor group (p < 0.05). In the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, the IL-6 inhibitor group had much higher apoptosis rates than the controls (p < 0.05). CONCLUSION Our findings indicate that inhibiting the IL-6/STAT3/HIF-1α signaling pathways could suppress IH growth.
Collapse
Affiliation(s)
- Aziguli Maimaiti
- Department of Pediatric Surgery, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yeerfan Aierken
- Department of Pediatric Surgery, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Department of Pediatric Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Ling Zhou
- Department of Pediatric Surgery, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Jun He
- Department of Pediatric Surgery, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Abudusaimi Abudureyimu
- Department of Pediatric Surgery, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Shui-Xue Li
- Department of Pediatric Surgery, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
18
|
Effects of Tocilizumab on Inflammation and Iron Metabolism in Critically Ill Patients with COVID-19. Pharmaceutics 2023; 15:pharmaceutics15020646. [PMID: 36839968 PMCID: PMC9960594 DOI: 10.3390/pharmaceutics15020646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/12/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
COVID-19 produces cytokine-mediated persistent inflammation and is associated with elevated iron stores and low circulating iron. It is believed that central to the pathophysiological mechanism is interleukin 6 and hepcidin. A state of iron overload, termed hyperferritinemia, and inflammatory anemia take place. Both conditions are linked to a worse result in critically ill patients. Blocking the interleukin 6-hepcidin pathway with Tocilizumab could present favorable outcomes. The aim of this study was to evaluate if Tocilizumab influences survival, the occurrence of sepsis, anemia and transfusions in critically ill patients suffering from COVID-19. This prospective observational study focused on levels of interleukin 6, hepcidin and blood iron parameters in patients treated with Tocilizumab. Data were compared before and after therapy as well as between treated and control groups. Results indicate that there is no difference in terms of survival nor in the rate of anemia or sepsis occurrence. Hepcidin was elevated and anemia ensued after treatment, which could indicate alternative pathways. In conclusion, when the classic interleukin 6-hepcidin pathway is blocked, inflammation seems to use alternative routes. Further understanding of these pathways is required and new pharmacological therapies need to be developed to treat persistent inflammation.
Collapse
|
19
|
Zhang Z, Xu D, Wang J, Zhang R, Du H, Zhou T, Wang X, Wang F. Rolling Circle Amplification-Based DNA Nano-Assembly for Targeted Drug Delivery and Gene Therapy. Biomacromolecules 2023; 24:439-448. [PMID: 36473109 DOI: 10.1021/acs.biomac.2c01271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Combining the killing ability of chemotherapy drugs on tumor cells with the inhibiting ability of genetic drugs on tumor cell growth, a dual drug delivery system loaded with therapy drugs and siRNA has gradually received more and more research and extensive attention. In this paper, we designed a DNA nano-assembly based on rolling circle amplification that can co-deliver doxorubicin (Dox) and siRNA simultaneously. In order to fully exploit the potential of the dual loading system in cancer treatment, we selected STAT3 gene as a target and used siRNA to target STAT3 of mRNA and reduce the STAT3 expression in mouse melanoma cell line (B16); meanwhile, Dox as a chemotherapy drug was combined with multivalent aptamers specifically targeting B16 to achieve efficient delivery of siRNA and Dox. The results showed that the synergistic delivery system could achieve high efficiency in targeting and inhibiting proliferation in mouse melanoma cells. In addition, the synergistic effect of the dual delivery system on apoptosis of cancer cells was significantly better than that of single drug delivery systems.
Collapse
Affiliation(s)
- Zhiqing Zhang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Dongyan Xu
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Jiawei Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Ruyan Zhang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Huan Du
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Ting Zhou
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Xiufeng Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Fang Wang
- College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| |
Collapse
|
20
|
Sil S, Bertilla J, Rupachandra S. A comprehensive review on RNA interference-mediated targeting of interleukins and its potential therapeutic implications in colon cancer. 3 Biotech 2023; 13:18. [PMID: 36568500 PMCID: PMC9768089 DOI: 10.1007/s13205-022-03421-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Colon cancer is the world's fourth leading cause of death. It is cancer of the latter part of the large intestine, i.e. the colon. Chronic inflammation over a long period also leads to the development of cancer. Cancer in the colon region is arduous to diagnose and is detected at a later stage when it metastasizes to other parts of the body like the liver, lungs, peritoneum, etc. Colon cancer is a great example of solid tumours associated with chronic inflammation. Although conventional therapies are effective, they lose their effectiveness beyond a certain point. Relapse of the disease occurs frequently. RNA interference (RNAi) is emerging as a great tool to specifically attack the cancer cells of a target site like the colon. RNAi deals with epigenetic changes made in the defective cells which ultimately leads to their death without harming the healthy cells. In this review, two types of epigenetic modulators have been considered, namely siRNA and miRNA, and their effect on interleukins. Interleukins, a class of cytokines, are major inflammatory responses of the body that are released by immune cells like leukocytes and macrophages. Some of these interleukins are pro-inflammatory, thereby promoting inflammation which eventually causes cancer. RNAi can prevent colon cancer by inhibiting pro-inflammatory interleukins.
Collapse
Affiliation(s)
- Sagari Sil
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu 603 203 India
| | - Janet Bertilla
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu 603 203 India
| | - S. Rupachandra
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu 603 203 India
| |
Collapse
|
21
|
Le HV, Le Cerf D. Colloidal Polyelectrolyte Complexes from Hyaluronic Acid: Preparation and Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2204283. [PMID: 36260830 DOI: 10.1002/smll.202204283] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/14/2022] [Indexed: 06/16/2023]
Abstract
Hyaluronic acid (HA) is a naturally occurring polysaccharide which has been extensively exploited in biomedical fields owing to its outstanding biocompatibility. Self-assembly of HA and polycations through electrostatic interactions can generate colloidal polyelectrolyte complexes (PECs), which can offer a wide range of applications while being relatively simple to prepare with rapid and "green" processes. The advantages of colloidal HA-based PECs stem from the combined benefits of nanomedicine, green chemistry, and the inherent properties of HA, namely high biocompatibility, biodegradability, and biological targeting capability. Accordingly, colloidal PECs from HA have received increasing attention in the recent years as high-performance materials for biomedical applications. Considering their potential, this review is aimed to provide a comprehensive understanding of colloidal PECs from HA in complex with polycations, from the most fundamental aspects of the preparation process to their various biomedical applications, notably as nanocarriers for delivering small molecule drugs, nucleic acids, peptides, proteins, and bioimaging agents or the construction of multifunctional platforms.
Collapse
Affiliation(s)
- Huu Van Le
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS UMR 6270, Rouen, 76000, France
| | - Didier Le Cerf
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS UMR 6270, Rouen, 76000, France
| |
Collapse
|
22
|
Sharma VK, Liu X, Oyarzún DA, Abdel-Azeem AM, Atanasov AG, Hesham AEL, Barik SK, Gupta VK, Singh BN. Microbial polysaccharides: An emerging family of natural biomaterials for cancer therapy and diagnostics. Semin Cancer Biol 2022; 86:706-731. [PMID: 34062265 DOI: 10.1016/j.semcancer.2021.05.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/07/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023]
Abstract
Microbial polysaccharides (MPs) offer immense diversity in structural and functional properties. They are extensively used in advance biomedical science owing to their superior biodegradability, hemocompatibility, and capability to imitate the natural extracellular matrix microenvironment. Ease in tailoring, inherent bio-activity, distinct mucoadhesiveness, ability to absorb hydrophobic drugs, and plentiful availability of MPs make them prolific green biomaterials to overcome the significant constraints of cancer chemotherapeutics. Many studies have demonstrated their application to obstruct tumor development and extend survival through immune activation, apoptosis induction, and cell cycle arrest by MPs. Synoptic investigations of MPs are compulsory to decode applied basics in recent inclinations towards cancer regimens. The current review focuses on the anticancer properties of commercially available and newly explored MPs, and outlines their direct and indirect mode of action. The review also highlights cutting-edge MPs-based drug delivery systems to augment the specificity and efficiency of available chemotherapeutics, as well as their emerging role in theranostics.
Collapse
Affiliation(s)
- Vivek K Sharma
- Pharmacology Division, CSIR-National Botanical Research Institute, Lucknow 226001, India
| | - Xiaowen Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Xuhui, Shanghai 200032, China.
| | - Diego A Oyarzún
- School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom; School of Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Ahmed M Abdel-Azeem
- Botany and Microbiology Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Atanas G Atanasov
- Institute for Digital Health and Patient Safety, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria; Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland; Institute of Neurobiology, Bulgarian Academy of Sciences, 23 Acad. G. Bonchev Str., 1113 Sofia, Bulgaria; Department of Pharmacognosy, University of Vienna, Althanstraße 14, 1090 Vienna, Austria
| | - Abd El-Latif Hesham
- Genetics Department, Faculty of Agriculture, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Saroj K Barik
- Pharmacology Division, CSIR-National Botanical Research Institute, Lucknow 226001, India
| | - Vijai Kumar Gupta
- Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, United Kingdom; Center for Safe and Improved Food, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh, EH9 3JG, United Kingdom.
| | - Brahma N Singh
- Pharmacology Division, CSIR-National Botanical Research Institute, Lucknow 226001, India.
| |
Collapse
|
23
|
Wu S, Liu C, Bai S, Lu Z, Liu G. Broadening the Horizons of RNA Delivery Strategies in Cancer Therapy. Bioengineering (Basel) 2022; 9:576. [PMID: 36290544 PMCID: PMC9598637 DOI: 10.3390/bioengineering9100576] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/05/2022] [Accepted: 10/11/2022] [Indexed: 12/02/2022] Open
Abstract
RNA-based therapy is a promising and innovative strategy for cancer treatment. However, poor stability, immunogenicity, low cellular uptake rate, and difficulty in endosomal escape are considered the major obstacles in the cancer therapy process, severely limiting the development of clinical translation and application. For efficient and safe transport of RNA into cancer cells, it usually needs to be packaged in appropriate carriers so that it can be taken up by the target cells and then be released to the specific location to perform its function. In this review, we will focus on up-to-date insights of the RNA-based delivery carrier and comprehensively describe its application in cancer therapy. We briefly discuss delivery obstacles in RNA-mediated cancer therapy and summarize the advantages and disadvantages of different carriers (cationic polymers, inorganic nanoparticles, lipids, etc.). In addition, we further summarize and discuss the current RNA therapeutic strategies approved for clinical use. A comprehensive overview of various carriers and emerging delivery strategies for RNA delivery, as well as the current status of clinical applications and practice of RNA medicines are classified and integrated to inspire fresh ideas and breakthroughs.
Collapse
Affiliation(s)
- Shuaiying Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Chao Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Shuang Bai
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhixiang Lu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| |
Collapse
|
24
|
Adibfar S, Masjedi A, Nazer A, Rashidi B, Karpisheh V, Izadi S, Hassannia H, Gholizadeh Navashenaq J, Mohammadi H, Hojjat-Farsangi M, Tarokhian H, Jadidi-Niaragh F. Combined inhibition of EZH2 and CD73 molecules by folic acid-conjugated SPION-TMC nanocarriers loaded with siRNA molecules prevents TNBC progression and restores anti-tumor responses. Life Sci 2022; 309:121008. [PMID: 36179812 DOI: 10.1016/j.lfs.2022.121008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/16/2022] [Accepted: 09/24/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Abnormal function or overexpression of CD73 and EZH2 within the tumor microenvironment and tumor cells enhances tumor growth and progression, and in many cases, causes drug resistance. Hence, it seems that silencing the expression of CD73 and EZH2 molecules in breast cancer reduces cancer development and enhances anti-tumor immune responses. METHODS we used siRNA-loaded superparamagnetic iron oxide (SPIONs) nanoparticles (NPs) coated with trimethyl chitosan (TMC) and functionalized with folic acid for co-delivery of EZH2/CD73 siRNAs to 4 T1 murine cancer cells both in vitro and in vivo. RESULTS Combination therapy markedly inhibited cancer cells' proliferation, migration, and viability and induced apoptosis in vitro. Moreover, in vivo administration of this combination therapy promoted tumor regression and induced anti-tumor immune responses. DISCUSSION The findings indicated the CD73/EZH2 factors inhibition by SPION-TMC-FA NPs as a promising therapeutic strategy in breast cancer treatment.
Collapse
Affiliation(s)
- Sara Adibfar
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran; Student Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Masjedi
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, Munich 81675, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich 81675, Germany
| | - Atefeh Nazer
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bentolhoda Rashidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Karpisheh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Izadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Hassannia
- Immunogenetic Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Hamed Mohammadi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; Department of Immunology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Hanieh Tarokhian
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
25
|
Barshidi A, Karpisheh V, Noukabadi FK, Kiani FK, Mohammadi M, Afsharimanesh N, Ebrahimi F, Kiaie SH, Navashenaq JG, Hojjat-Farsangi M, Zolbanin NM, Mahmoodpoor A, Hassannia H, Nami S, Jalali P, Jafari R, Jadidi-Niaragh F. Dual Blockade of PD-1 and LAG3 Immune Checkpoints Increases Dendritic Cell Vaccine Mediated T Cell Responses in Breast Cancer Model. Pharm Res 2022; 39:1851-1866. [PMID: 35715669 DOI: 10.1007/s11095-022-03297-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Increasing the efficiency of unsuccessful immunotherapy methods is one of the most important research fields. Therefore, the use of combination therapy is considered as one of the ways to increase the effectiveness of the dendritic cell (DC) vaccine. In this study, the inhibition of immune checkpoint receptors such as LAG3 and PD-1 on T cells was investigated to increase the efficiency of T cells in response to the DC vaccine. METHODS We used trimethyl chitosan-dextran sulfate-lactate (TMC-DS-L) nanoparticles (NPs) loaded with siRNA molecules to quench the PD-1 and LAG3 checkpoints' expression. RESULTS Appropriate physicochemical characteristics of the generated NPs led to efficient inhibition of LAG3 and PD-1 on T cells, which was associated with increased survival and activity of T cells, ex vivo. Also, treating mice with established breast tumors (4T1) using NPs loaded with siRNA molecules in combination with DC vaccine pulsed with tumor lysate significantly inhibited tumor growth and increased survival in mice. These ameliorative effects were associated with increased anti-tumor T cell responses and downregulation of immunosuppressive cells in the tumor microenvironment and spleen. CONCLUSION These findings strongly suggest that TMC-DS-L NPs loaded with siRNA could act as a novel tool in inhibiting the expression of immune checkpoints in the tumor microenvironment. Also, combination therapy based on inhibition of PD-1 and LAG3 in combination with DC vaccine is an effective method in treating cancer that needs to be further studied.
Collapse
Affiliation(s)
- Asal Barshidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Karpisheh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Fariba Karoon Kiani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Mohammadi
- Department of Cell and Molecular Biology, School of Advanced Sciences, Islamic Azad University, Tehran Medical Branch, Tehran, , Iran
| | - Negin Afsharimanesh
- Department of Microbiology, Faculty of Basic Sciences, Hamedan Branch, Islamic Azad University, Hamedan, Iran
| | - Farbod Ebrahimi
- Nanoparticle Process Technology, Faculty of Engineering, University of Duisburg-Essen, Duisburg, Germany
| | - Seyed Hossein Kiaie
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- The Persian Gulf Marine Biotechnology Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Naime Majidi Zolbanin
- Experimental and Applied Pharmaceutical Sciences Research Center,, Urmia University of Medical Sciences, Urmia, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, , Urmia University of Medical Sciences, Urmia, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, School of Medicine, Imam Reza Medical Research & Training Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Hassannia
- Immunogenetic Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sanam Nami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pooya Jalali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
26
|
Functionalized chitosan as a promising platform for cancer immunotherapy: A review. Carbohydr Polym 2022; 290:119452. [DOI: 10.1016/j.carbpol.2022.119452] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/20/2022]
|
27
|
Ahmad MZ, Alasiri AS, Alasmary MY, Abdullah MM, Ahmad J, Abdel Wahab BA, M Alqahtani SA, Pathak K, Mustafa G, Khan MA, Saikia R, Gogoi U. Emerging advances in nanomedicine for breast cancer immunotherapy: opportunities and challenges. Immunotherapy 2022; 14:957-983. [PMID: 35852105 DOI: 10.2217/imt-2021-0348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Breast cancer is one of the most common causes of cancer-related morbidity and mortality in women worldwide. Early diagnosis and an appropriate therapeutic approach for all cancers are climacterics for a favorable prognosis. Targeting the immune system in breast cancer is already a clinical reality with notable successes, specifically with checkpoint blockade antibodies and chimeric antigen receptor T-cell therapy. However, there have been inevitable setbacks in the clinical application of cancer immunotherapy, including inadequate immune responses due to insufficient delivery of immunostimulants to immune cells and uncontrolled immune system modulation. Rapid advancements and new evidence have suggested that nanomedicine-based immunotherapy may be a viable option for treating breast cancer.
Collapse
Affiliation(s)
- Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia
| | - Ali S Alasiri
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia
| | - Mohammed Yahia Alasmary
- Medical Department, College of Medicine, Najran University, Najran, 11001, Kingdom of Saudi Arabia
| | - M M Abdullah
- Advanced Materials & Nano-Research Centre, Department of Physics, Faculty of Science & Arts, Najran University, Najran, 11001, Kingdom Saudi Arabia
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia
| | - Basel A Abdel Wahab
- Department of Pharmacology, College of Pharmacy, Najran University, Najran, 11001, Kingdom of Saudi Arabia
- Department of Pharmacology, College of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Saif Aboud M Alqahtani
- Internal Medicine Department, College of Medicine, King Khalid University, Abha, 61421, Kingdom of Saudi Arabia
| | - Kalyani Pathak
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Gulam Mustafa
- College of Pharmacy, Shaqra University, Ad-Dawadmi Riyadh, Kingdom of Saudi Arabia
| | - Mohammad Ahmad Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Riya Saikia
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Urvashee Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India
| |
Collapse
|
28
|
Vetter VC, Wagner E. Targeting nucleic acid-based therapeutics to tumors: Challenges and strategies for polyplexes. J Control Release 2022; 346:110-135. [PMID: 35436520 DOI: 10.1016/j.jconrel.2022.04.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/07/2022] [Accepted: 04/10/2022] [Indexed: 12/18/2022]
Abstract
The current medical reality of cancer gene therapy is reflected by more than ten approved products on the global market, including oncolytic and other viral vectors and CAR T-cells as ex vivo gene-modified cell therapeutics. The development of synthetic antitumoral nucleic acid therapeutics has been proceeding at a lower but steady pace, fueled by a plethora of alternative nucleic acid platforms (from various antisense oligonucleotides, siRNA, microRNA, lncRNA, sgRNA, to larger mRNA and DNA) and several classes of physical and chemical delivery technologies. This review summarizes the challenges and strategies for tumor-targeted nucleic acid delivery. Focusing primarily on polyplexes (polycation complexes) as nanocarriers, delivery options across multiple barriers into tumor cells are illustrated.
Collapse
Affiliation(s)
- Victoria C Vetter
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig-Maximilians-Universität, Munich 81377, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig-Maximilians-Universität, Munich 81377, Germany; Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Munich 81377, Germany.
| |
Collapse
|
29
|
Shang M, Wu Y, Wang Y, Cai Y, Jin J, Yang Z. Dual antisense oligonucleotide targeting miR-21/miR-155 synergize photodynamic therapy to treat triple-negative breast cancer and inhibit metastasis. Biomed Pharmacother 2022; 146:112564. [PMID: 34954643 DOI: 10.1016/j.biopha.2021.112564] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/12/2021] [Accepted: 12/19/2021] [Indexed: 12/14/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a greatly aggressive subtype of breast cancer with high recurrence and mortality rates. Chemotherapy as a primary treatment for cancer is limited due to toxic side effects and drug resistance. Therefore, low toxicity and more effective breast cancer therapeutic approaches are greatly desired. In this study, a strategy which using ZIF-90 nanoparticles co-deliver Ce6-anti-miR-21 and Ce6-anti-miR-155 into the tumor cells was developed. Due to the pH responsive drug release of ZIF-90, antisense oligonucleotides (anti-miRNAs) and photosensitizers are able to be efficiently released inside tumor microenvironment. The nano delivery system captures overexpressed oncogenic miRNAs while the photosensitizer Ce6 generates ROS under light irradiation to effectively induce the apoptosis of tumor cell. This combinatorial effect was verified by results showing that the purposed therapic method could effectively inhibit tumor cell proliferation and metastasis. The concept of antisense oligonucleotide combined with photodynamic therapy has great potential in cancer treatment or adjuvant therapy.
Collapse
Affiliation(s)
- Mengdi Shang
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Yiyang Wu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Yeyang Wang
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Yanfei Cai
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China
| | - Jian Jin
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China.
| | - Zhaoqi Yang
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
30
|
Karoon Kiani F, Izadi S, Ansari Dezfouli E, Ebrahimi F, Mohammadi M, Chalajour H, Mortazavi Bulus M, Nasr Esfahani M, Karpisheh V, Mahmoud Salehi Khesht A, Abbaszadeh-Goudarzi K, Soleimani A, Gholizadeh Navashenaq J, Ahmadi M, Hassannia H, Hojjat-Farsangi M, Shahmohammadi Farid S, Hashemi V, Jadidi-Niaragh F. Simultaneous silencing of the A2aR and PD-1 immune checkpoints by siRNA-loaded nanoparticles enhances the immunotherapeutic potential of dendritic cell vaccine in tumor experimental models. Life Sci 2022; 288:120166. [PMID: 34813798 DOI: 10.1016/j.lfs.2021.120166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 12/20/2022]
Abstract
Following various immunotherapies, lack of proper anti-tumor immune responses is considered a significant problem in novel cancer therapeutic approaches. The expression of inhibitory checkpoint molecules on tumor-infiltrating T cells is one of the main reasons for the ineffectiveness of various immunotherapies. Therefore, we decided to inhibit two of the most important immune checkpoints expressed on tumor-associated T cells, PD-1 and A2aR. Ligation of PD-1 with PD-L1 and A2aR with adenosine significantly suppress T cell responses against tumor cells. Whitin tumors, specific inhibition of these molecules on T cells is of particular importance for successful immunotherapy as well as the elimination of treatment-associated side-effects. Thus, in this study, superparamagnetic iron oxide (SPION) nanoparticles (NPs) were covered by chitosan lactate (CL), functionalized with TAT peptide, and loaded with siRNA molecules against PD-1 and A2aR. Appropriate physicochemical properties of the prepared NPs resulted in efficient delivery of siRNA to tumor-derived T cells and suppressed the expression of A2aR and PD-1, ex vivo. T cell functions such as cytokine secretion and proliferation were considerably enhanced by the downregulation of these molecules which led to an increase in their survival time. Interestingly, treatment of CT26 and 4T1 mouse tumors with siRNA-loaded NPs not only inhibited tumor growth but also markedly increased anti-tumor immune responses and survival time. The results strongly support the efficacy of SPION-CL-TAT NPs loaded with anti-PD-1/A2aR siRNAs in cancer therapy and their further development for cancer patients in the near future.
Collapse
Affiliation(s)
- Fariba Karoon Kiani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Izadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ehsan Ansari Dezfouli
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farbod Ebrahimi
- Nanoparticle Process Technology, Faculty of Engineering, University of Duisburg-Essen, Duisburg, Germany
| | - Mohammad Mohammadi
- Department of Cell and Molecular Biology, School of Advanced Sciences, Islamic Azad University, Tehran, Medical Branch, Iran
| | - Hengameh Chalajour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Vahid Karpisheh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Armin Mahmoud Salehi Khesht
- Department of Biochemistry, Faculty of Materials Engineering, Islamic Azad University, Najafabad Branch, Najafabad, Iran
| | | | - Ali Soleimani
- Department of Public Health, Maragheh University of Medical Sciences, Maragheh, Iran
| | | | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Hassannia
- Immunogenetic Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden; Department of Immunology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | | | - Vida Hashemi
- Department of Basic Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
31
|
Fathi M, Bahmanpour S, Barshidi A, Rasouli H, Karoon Kiani F, Mahmoud Salehi Khesht A, Izadi S, Rashidi B, Kermanpour S, Mokhtarian R, Karpisheh V, Hassannia H, Mohammadi H, Jalili A, Jadidi-Niaragh F. Simultaneous blockade of TIGIT and HIF-1α induces synergistic anti-tumor effect and decreases the growth and development of cancer cells. Int Immunopharmacol 2021; 101:108288. [PMID: 34710844 DOI: 10.1016/j.intimp.2021.108288] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 01/19/2023]
Abstract
PURPOSE T-cell immunoglobulin and ITIM domain (TIGIT) is an immune checkpoint that is overexpressed on both immune cells and some cancer cells. TIGIT can alter the anti-tumor responses inside the tumor microenvironment. Hypoxia-inducible factor 1-alpha (HIF-1α) plays a significant role in the TME and involves suppressing the anti-tumor responses. Under hypoxic conditions, HIF-1α can enhance the expression of different immune checkpoints. Accordingly, hypoxic TME and TIGIT overexpression cause cancer development. Thus, we decided to inhibit tumor cell expansion by inhibiting TIGIT and HIF-1α molecules and discovering the relationship between TIGIT and HIF-1α. METHODS In this research, we utilized superparamagnetic iron oxide-based NPs (SPIONs) combined with chitosan lactate (CL) and folic acid (FA) nanoparticles (NPs) loaded with TIGIT-siRNA and HIF-1α- siRNA for suppressing TIGIT and HIF-1α in tumor cells and evaluated the consequences of this treatment strategy on tumor growth, apoptosis, and metastasis. RESULTS The results showed that cancer cells treated with TIGIT and HIF-1α siRNA-loaded SPIONs-CL-FA NPs, strongly suppressed the TIGIT and HIF-1α expression, colony formation ability, angiogenesis, and the growth rate of cancer cells. CONCLUSIONS Present data suggest the combination treatment of TIGIT and HIF-1α as a novel treatment strategy against colorectal and breast cancer, but more researches are required to realize the complete role of TIGIT and HIF-1α inside the TME.
Collapse
Affiliation(s)
- Mehrdad Fathi
- Student Research Committee, Kurdistan University of Medical Sciences, Sanandaj, Iran; Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran; Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Simin Bahmanpour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asal Barshidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Rasouli
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Karoon Kiani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sepideh Izadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bentolhoda Rashidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Kermanpour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roya Mokhtarian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Karpisheh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Hassannia
- Immunogenetic Research Center, Faculty of Medicine and Amol Faculty of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hamed Mohammadi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Ali Jalili
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran; Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
32
|
Therapeutic approaches targeting molecular signaling pathways common to diabetes, lung diseases and cancer. Adv Drug Deliv Rev 2021; 178:113918. [PMID: 34375681 DOI: 10.1016/j.addr.2021.113918] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/23/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus (DM), is the most common metabolic disease and is characterized by sustained hyperglycemia. Accumulating evidences supports a strong association between DM and numerous lung diseases including chronic obstructive pulmonary disease (COPD), fibrosis, and lung cancer (LC). The global incidence of DM-associated lung disorders is rising and several ongoing studies, including clinical trials, aim to elucidate the molecular mechanisms linking DM with lung disorders, in particular LC. Several potential mechanisms, including hyperglycemia, hyperinsulinemia, glycation, inflammation, and hypoxia, are cited as plausible links between DM and LC. In addition, studies also propose a connection between the use of anti-diabetic medications and reduction in the incidence of LC. However, the exact cause for DM associated lung diseases especially LC is not clear and is an area under intense investigation. Herein, we review the biological links reported between DM and lung disorders with an emphasis on LC. Furthermore, we report common signaling pathways (eg: TGF-β, IL-6, HIF-1, PDGF) and miRNAs that are dysregulated in DM and LC and serve as molecular targets for therapy. Finally, we propose a nanomedicine based approach for delivering therapeutics (eg: IL-24 plasmid DNA, HuR siRNA) to disrupt signaling pathways common to DM and LC and thus potentially treat DM-associated LC. Finally, we conclude that the effective modulation of commonly regulated signaling pathways would help design novel therapeutic protocols for treating DM patients diagnosed with LC.
Collapse
|
33
|
Della Sala F, Fabozzi A, di Gennaro M, Nuzzo S, Makvandi P, Solimando N, Pagliuca M, Borzacchiello A. Advances in Hyaluronic-Acid-Based (Nano)Devices for Cancer Therapy. Macromol Biosci 2021; 22:e2100304. [PMID: 34657388 DOI: 10.1002/mabi.202100304] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/08/2021] [Indexed: 12/12/2022]
Abstract
Cancer is the main cause of fatality all over the world with a considerable growth rate. Many biologically active nanoplatforms are exploited for tumor treatment. Of nanodevices, hyaluronic acid (HA)-based systems have shown to be promising candidates for cancer therapy due to their high biocompatibility and cell internalization. Herein, surface functionalization of different nanoparticles (NPs), e.g., organic- and inorganic-based NPs, is highlighted. Subsequently, HA-based nanostructures and their applications in cancer therapy are presented.
Collapse
Affiliation(s)
- Francesca Della Sala
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Antonio Fabozzi
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Mario di Gennaro
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Stefano Nuzzo
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Pooyan Makvandi
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Nicola Solimando
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Maurizio Pagliuca
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Assunta Borzacchiello
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| |
Collapse
|
34
|
Application of Non-Viral Vectors in Drug Delivery and Gene Therapy. Polymers (Basel) 2021; 13:polym13193307. [PMID: 34641123 PMCID: PMC8512075 DOI: 10.3390/polym13193307] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/15/2021] [Accepted: 09/18/2021] [Indexed: 12/13/2022] Open
Abstract
Vectors and carriers play an indispensable role in gene therapy and drug delivery. Non-viral vectors are widely developed and applied in clinical practice due to their low immunogenicity, good biocompatibility, easy synthesis and modification, and low cost of production. This review summarized a variety of non-viral vectors and carriers including polymers, liposomes, gold nanoparticles, mesoporous silica nanoparticles and carbon nanotubes from the aspects of physicochemical characteristics, synthesis methods, functional modifications, and research applications. Notably, non-viral vectors can enhance the absorption of cargos, prolong the circulation time, improve therapeutic effects, and provide targeted delivery. Additional studies focused on recent innovation of novel synthesis techniques for vector materials. We also elaborated on the problems and future research directions in the development of non-viral vectors, which provided a theoretical basis for their broad applications.
Collapse
|
35
|
Mikušová V, Mikuš P. Advances in Chitosan-Based Nanoparticles for Drug Delivery. Int J Mol Sci 2021; 22:9652. [PMID: 34502560 PMCID: PMC8431817 DOI: 10.3390/ijms22179652] [Citation(s) in RCA: 224] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/17/2022] Open
Abstract
Nanoparticles (NPs) have an outstanding position in pharmaceutical, biological, and medical disciplines. Polymeric NPs based on chitosan (CS) can act as excellent drug carriers because of some intrinsic beneficial properties including biocompatibility, biodegradability, non-toxicity, bioactivity, easy preparation, and targeting specificity. Drug transport and release from CS-based particulate systems depend on the extent of cross-linking, morphology, size, and density of the particulate system, as well as physicochemical properties of the drug. All these aspects have to be considered when developing new CS-based NPs as potential drug delivery systems. This comprehensive review is summarizing and discussing recent advances in CS-based NPs being developed and examined for drug delivery. From this point of view, an enhancement of CS properties by its modification is presented. An enhancement in drug delivery by CS NPs is discussed in detail focusing on (i) a brief summarization of basic characteristics of CS NPs, (ii) a categorization of preparation procedures used for CS NPs involving also recent improvements in production schemes of conventional as well as novel CS NPs, (iii) a categorization and evaluation of CS-based-nanocomposites involving their production schemes with organic polymers and inorganic material, and (iv) very recent implementations of CS NPs and nanocomposites in drug delivery.
Collapse
Affiliation(s)
- Veronika Mikušová
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia;
| | - Peter Mikuš
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia
| |
Collapse
|
36
|
Mahmoud Salehi Khesht A, Karpisheh V, Qubais Saeed B, Olegovna Zekiy A, Yapanto LM, Nabi Afjadi M, Aksoun M, Nasr Esfahani M, Aghakhani F, Movahed M, Joshi N, Abbaszadeh-Goudarzi K, Hallaj S, Ahmadi M, Dolati S, Mahmoodpoor A, Hashemi V, Jadidi-Niaragh F. Different T cell related immunological profiles in COVID-19 patients compared to healthy controls. Int Immunopharmacol 2021; 97:107828. [PMID: 34091116 PMCID: PMC8162824 DOI: 10.1016/j.intimp.2021.107828] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022]
Abstract
In various pathological conditions, cellular immunity plays an important role in immune responses. Amongimmunecells, T lymphocytes pdomotecellular and humoralresponses as well as innate immunity. Therefore, careful investigation of these cells has a significant impact on accurate knowledge in COVID-19diseasepathogenesis. In current research, the frequency and function of various T lymphocytes involved in immune responses examined in SARS-CoV-2 patients with various disease severity compared to normal subjects. In order to make an accurate comparison among patients with various disease severity, this study was performed on asymptomatic recovered cases (n = 20), ICU hospitalized patients (n = 30), non-ICU hospitalized patients (n = 30), and normal subjects (n = 20). To precisely evaluate T cells activity following purification, their cytokine secretion activity was examined. Similarly, immediately after purification of Treg cells, their inhibitory activity on T cells was investigated. The results showed that COVID-19 patients with severe disease (ICU hospitalized patients) not only had a remarkable increase in Th1 and Th17 but also a considerable decrease in Th2 and Treg cells. More importantly, as the IL-17 and IFN-γ secretion was sharply increased in severe disease, the secretion of IL-10 and IL-4 was decreased. Furthermore, the inhibitory activity of Treg cells was reduced in severe disease patients in comparison to other groups. In severe COVID-19 disease, current findings indicate when the inflammatory arm of cellular immunity is significantly increased, a considerable reduction in anti-inflammatory and regulatory arm occurred.
Collapse
Affiliation(s)
- Armin Mahmoud Salehi Khesht
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Materials Engineering, Islamic Azad University, Najafabad Branch, Najafabad, Iran
| | - Vahid Karpisheh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Balsam Qubais Saeed
- Clinical Sciences Department, College of Medicine, University of Sharjah, United Arab Emirates
| | - Angelina Olegovna Zekiy
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Lis M Yapanto
- Department of Aquatic Management, Faculty of Fisheries and Marine Science Universitas Negeri Gorontalo, Gorontalo, Indonesia
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, University of Tarbiat Modares, Tehran, Iran
| | - Mohsen Aksoun
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Fatemeh Aghakhani
- Department of Microbiology, Tehran Medical Branch, Islamic Azad University, Tehran, Iran
| | - Mahsa Movahed
- Department of Biology, Faculty of Sciences, Yazd University, Yazd, Iran
| | - Navneet Joshi
- Department of Biosciences, Mody University of Science and Technology, Lakshmangarh, Rajasthan, India
| | - Kazem Abbaszadeh-Goudarzi
- Department of Medical Biotechnology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Shahin Hallaj
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Faculty of Medicine, Imam Reza Medical Research & Training Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vida Hashemi
- Department of Basic Science, Faculty of Medicine, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
37
|
Mapanao AK, Che PP, Sarogni P, Sminia P, Giovannetti E, Voliani V. Tumor grafted - chick chorioallantoic membrane as an alternative model for biological cancer research and conventional/nanomaterial-based theranostics evaluation. Expert Opin Drug Metab Toxicol 2021; 17:947-968. [PMID: 33565346 DOI: 10.1080/17425255.2021.1879047] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/18/2021] [Indexed: 02/09/2023]
Abstract
Introduction: Advancements in cancer management and treatment are associated with strong preclinical research data, in which reliable cancer models are demanded. Indeed, inconsistent preclinical findings and stringent regulations following the 3Rs principle of reduction, refinement, and replacement of conventional animal models currently pose challenges in the development and translation of efficient technologies. The chick embryo chorioallantoic membrane (CAM) is a system for the evaluation of treatment effects on the vasculature, therefore suitable for studies on angiogenesis. Apart from vascular effects, the model is now increasingly employed as a preclinical cancer model following tumor-grafting procedures.Areas covered: The broad application of CAM tumor model is highlighted along with the methods for analyzing the neoplasm and vascular system. The presented and cited investigations focus on cancer biology and treatment, encompassing both conventional and emerging nanomaterial-based modalities.Expert opinion: The CAM tumor model finds increased significance given the influences of angiogenesis and the tumor microenvironment in cancer behavior, then providing a qualified miniature system for oncological research. Ultimately, the establishment and increased employment of such a model may resolve some of the limitations present in the standard preclinical tumor models, thereby redefining the preclinical research workflow.
Collapse
Affiliation(s)
- Ana Katrina Mapanao
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
- NEST-Scuola Normale Superiore, Pisa, Italy
| | - Pei Pei Che
- Department of Radiation Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, The Netherlands
| | - Patrizia Sarogni
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
| | - Peter Sminia
- Department of Radiation Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per La Scienza, Pisa, Italy
| | - Valerio Voliani
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
| |
Collapse
|
38
|
Yu Y, Ren KM, Chen XL. Expression and role of P-element-induced wimpy testis-interacting RNA in diabetic-retinopathy in mice. World J Diabetes 2021; 12:1116-1130. [PMID: 34326959 PMCID: PMC8311480 DOI: 10.4239/wjd.v12.i7.1116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/11/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND As one of the major microvascular complications of diabetes, diabetic retinopathy (DR) is the leading cause of blindness in the working age population. Because the extremely complex pathogenesis of DR has not been fully clarified, the occurrence and development of DR is closely related to tissue ischemia and hypoxia and neovascularization The formation of retinal neovascularization (RNV) has great harm to the visual acuity of patients.
AIM To investigate the expression of P-element-induced wimpy testis-interacting RNA (piRNA) in proliferative DR mice and select piRNA related to RNV.
METHODS One hundred healthy C57BL/6J mice were randomly divided into a normal group as control group (CG) and proliferative DR (PDR) group as experimental group (EG), with 50 mice in each group. Samples were collected from both groups at the same time, and the lesions of mice were evaluated by hematoxylin and eosin staining and retinal blood vessel staining. The retinal tissues were collected for second-generation high-throughput sequencing, and the differentially expressed piRNA between the CG and EG was detected, and polymerase chain reaction (PCR) was conducted for verification. The differentially obtained piRNA target genes and expression profiles were enrichment analysis based on gene annotation (Gene Ontology) and Kyoto Encyclopedia of Genes and Genomes.
RESULTS In the CG there was no perfusion area, neovascularization and endothelial nucleus broke through the inner boundary membrane of retinap. In the EG, there were a lot of nonperfused areas, new blood vessels and endothelial nuclei breaking through the inner boundary membrane of the retina. There was a statistically significant difference in the number of vascular endothelial nuclei breaking through the inner retinal membrane between the two groups. High-throughput sequencing analysis showed that compared with the CG, a total of 79 piRNAs were differentially expressed in EG, among which 43 piRNAs were up-regulated and 36 piRNAs were down-regulated. Bioinformatics analysis showed that the differentially expressed piRNAs were mainly concentrated in the signaling pathways of angiogenesis and cell proliferation. Ten piRNAs were selected for PCR, and the results showed that the expression of piR-MMU-40373735, piR-MMU-61121420, piR-MMU-55687822, piR-MMU-1373887 were high, and the expression of piR-MMU-7401535, piR-MMU-4773779, piR-MMU-1304999, and piR-MMU-5160126 were low, which were consistent with the sequencing results.
CONCLUSION In the EG, the abnormal expression of piRNA is involved in the pathway of angiogenesis and cell proliferation, suggesting that piRNAs have some regulatory function in proliferative diabetic-retinopathy.
Collapse
Affiliation(s)
- Yong Yu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Kai-Ming Ren
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Xiao-Long Chen
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| |
Collapse
|
39
|
|
40
|
Zeinali M, Abbaspour-Ravasjani S, Soltanfam T, Paiva-Santos AC, Babaei H, Veiga F, Hamishehkar H. Prevention of UV-induced skin cancer in mice by gamma oryzanol-loaded nanoethosomes. Life Sci 2021; 283:119759. [PMID: 34171381 DOI: 10.1016/j.lfs.2021.119759] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 01/05/2023]
Abstract
AIMS Skin cancer is the most widespread cancer worldwide, mainly caused by exposure to ultraviolet radiation (UV) in sunlight. Utilizing topical preventive agents in routinely daily used cosmetics may prevent UV-related skin damages and skin cancers. γ-Oryzanol (GO) is a natural component derived from rice bran oil, with potential antioxidant and skin anti-aging properties. MAIN METHODS We biologically thorough studied the antioxidant and anticancer effects of GO in vitro to found the effective signaling pathways, then evaluated the sun protection factor of prepared formulation, and finally investigated the long-term preventive effects of GO-loaded nanoethosomes (GO-NEs) against UVB-induced skin cancer in mice. KEY FINDINGS GO-NEs could effectively prevent UVB-induced skin cancer. SIGNIFICANCE Our results suggest that GO-NEs could be utilized as an innovative ingredient in cosmetics.
Collapse
Affiliation(s)
- Mahdi Zeinali
- Biotechnology Research Center, Student Research Committee, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Tannaz Soltanfam
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Research and Development Unit, Daana Pharma. Co. Tabriz, Iran.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal.
| | - Hossein Babaei
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, Portugal.
| | - Hamed Hamishehkar
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
41
|
Allahyari SE, Hajizadeh F, Zekiy AO, Mansouri N, Gilan PS, Mousavi SM, Masjedi A, Hassannia H, Ahmadi M, Mohammadi H, Yousefi M, Izadi S, Zolbanin NM, Jafari R, Jadidi-Niaragh F. Simultaneous inhibition of CD73 and IL-6 molecules by siRNA-loaded nanoparticles prevents the growth and spread of cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 34:102384. [PMID: 33771704 DOI: 10.1016/j.nano.2021.102384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/11/2021] [Accepted: 03/03/2021] [Indexed: 01/11/2023]
Abstract
High concentrations of adenosine and interleukin (IL)-6 in the tumor microenvironment have been identified as one of the leading causes of cancer growth. Thus, we decided to inhibit the growth of cancer cells by inhibiting the production of adenosine and IL-6 in the tumor environment at the same time. For this purpose, we used chitosan-lactate-PEG-TAT (CLP-TAT) nanoparticles (NPs) loaded with siRNA molecules against CD73, an adenosine-producing enzyme, and IL-6. Proper physicochemical properties of the produced NPs led to high cell uptake and suppression of target molecules. Administration of these NPs to tumor-bearing mice (4T1 and CT26 models) greatly reduced the size of the tumor and increased the survival of the mice, which was accompanied by an increase in anti-tumor T lymphocyte responses. These findings suggest that combination therapy using siRNA-loaded CLP-TAT NPs against CD73 and IL-6 molecules could be an effective treatment strategy against cancer that needs further study.
Collapse
Affiliation(s)
- Sima Emadi Allahyari
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farnaz Hajizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Angelina Olegovna Zekiy
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Niloofar Mansouri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Sahami Gilan
- Medical Biology Research Center, Health Technologies Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Ali Masjedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Hassannia
- Immunogenetic Research Center, Faculty of Medicine and Amol Faculty of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Izadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naime Majidi Zolbanin
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Reza Jafari
- Solid Tumor Research Center, Cellular and Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
42
|
Joshi N, Hajizadeh F, Ansari Dezfouli E, Zekiy AO, Nabi Afjadi M, Mousavi SM, Hojjat-Farsangi M, Karpisheh V, Mahmoodpoor A, Hassannia H, Dolati S, Mohammadi H, Yousefi M, Jadidi-Niaragh F. Silencing STAT3 enhances sensitivity of cancer cells to doxorubicin and inhibits tumor progression. Life Sci 2021; 275:119369. [PMID: 33745894 DOI: 10.1016/j.lfs.2021.119369] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/21/2021] [Accepted: 03/07/2021] [Indexed: 12/15/2022]
Abstract
AIMS Despite extensive efforts to find new treatments, chemotherapy is still one of the first and foremost choices for cancer treatment. The main problems of using these drugs are the resistance of cancer cells and reducing their sensitivity to chemotherapy as well as the side effects of their systemic administration. Because STAT3 plays a very important role in the survival and susceptibility of cancer cells to apoptosis, we hypothesized that suppression of STAT3 expression could induce greater susceptibility to DOX-induced cancer cell death. MATERIALS AND METHODS We used pegylated chitosan lactate nanoparticles (NPs) functionalized by TAT peptide and folate to deliver STAT3 siRNA and DOX to cancer cells simultaneously, both in vitro and in vivo. KEY FINDINGS The results showed that NPs could effectively deliver siRNA and DOX to cancer cells, which was associated with suppression of STAT3 expression and increased induction of DOX-mediated cell death. Concomitant delivery of DOX and STAT3 siRNA also suppressed tumor growth in 4T1 and CT26 cancer models, which was associated with induction of anti-tumor immune responses. SIGNIFICANCE These findings suggest that the use of NPs can be an effective strategy for the targeted delivery of STAT3-specific siRNA/DOX to cancer cells.
Collapse
Affiliation(s)
- Navneet Joshi
- Department of Biosciences, Mody University of Science and Technology, Lakshmangarh, Rajasthan, India.
| | - Farnaz Hajizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ehsan Ansari Dezfouli
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Angelina Olegovna Zekiy
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, University of Tarbiat Modares, Tehran, Iran
| | | | | | - Vahid Karpisheh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, School of Medicine, Imam Reza Medical Research & Training Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Hassannia
- Immunogenetic Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
43
|
Budi HS, Izadi S, Timoshin A, Asl SH, Beyzai B, Ghaderpour A, Alian F, Eshaghi FS, Mousavi SM, Rafiee B, Nikkhoo A, Ahmadi A, Hassannia H, Ahmadi M, Sojoodi M, Jadidi-Niaragh F. Blockade of HIF-1α and STAT3 by hyaluronate-conjugated TAT-chitosan-SPION nanoparticles loaded with siRNA molecules prevents tumor growth. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 34:102373. [PMID: 33667724 DOI: 10.1016/j.nano.2021.102373] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/15/2020] [Accepted: 02/07/2021] [Indexed: 12/15/2022]
Abstract
HIF-1α and STAT3 are two of the critical factors in the growth, proliferation, and metastasis of cancer cells and play a crucial role in inhibiting anti-cancer immune responses. Therefore, we used superparamagnetic iron oxide (SPION) nanoparticles (NPs) coated with thiolated chitosan (ChT) and trimethyl chitosan (TMC) and functionalized with hyaluronate (H) and TAT peptide for delivery of siRNA molecules against STAT3 and HIF-1α to cancer cells both in vivo and in vitro. The results indicated that tumor cell transfection with siRNA-encapsulated NPs robustly inhibited proliferation and migration and induced apoptosis in tumor cells. Furthermore, simultaneous silencing of HIF-1α and STAT3 significantly repressed cancer development in two different tumor types (4T1 breast cancer and CT26 colon cancer) which were associated with upregulation of cytotoxic T lymphocytes and IFN-γ secretion. The findings suggest inhibiting the HIF-1α/STAT3 axis by SPION-TMC-ChT-TAT-H NPs as an effective way to treat cancer.
Collapse
Affiliation(s)
- Hendrik Setia Budi
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Sepideh Izadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Anton Timoshin
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Department of propaedeutics of dental diseases, Moscow, Russia
| | | | - Behzad Beyzai
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Ghaderpour
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Alian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Farzaneh Sadat Eshaghi
- Department of Genetics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Behnam Rafiee
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - Afshin Nikkhoo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Armin Ahmadi
- iepartment of Chemical and Materials Engineering, The University of Alabama in Huntsville, AL, USA
| | - Hadi Hassannia
- Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mozhdeh Sojoodi
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, USA
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
44
|
Brouillard A, Deshpande N, Kulkarni AA. Engineered Multifunctional Nano- and Biological Materials for Cancer Immunotherapy. Adv Healthc Mater 2021; 10:e2001680. [PMID: 33448159 DOI: 10.1002/adhm.202001680] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/21/2020] [Indexed: 12/19/2022]
Abstract
Cancer immunotherapy is set to emerge as the future of cancer therapy. However, recent immunotherapy trials in different cancers have yielded sub-optimal results, with durable responses seen in only a small fraction of patients. Engineered multifunctional nanomaterials and biological materials are versatile platforms that can elicit strong immune responses and improve anti-cancer efficacy when applied to cancer immunotherapy. While there are traditional systems such as polymer- and lipid-based nanoparticles, there is a wide variety of other materials with inherent and additive properties that can allow for more potent activation of the immune system. By synthesizing and applying multifunctional strategies, it allows for a more extensive and more effective repertoire of tools to use in the wide variety of situations that cancer presents itself. Here, several types of nanoscale and biological material strategies and platforms that provide their inherent benefits for targeting and activating multiple aspects of the immune system are discussed. Overall, this review aims to provide a comprehensive understanding of recent advances in the field of multifunctional cancer immunotherapy and trends that pave the way for more diverse and tactical regression of tumors through soliciting responses by either the adaptive or innate immune system, and even both simultaneously.
Collapse
Affiliation(s)
- Anthony Brouillard
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
| | - Nilesh Deshpande
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
| | - Ashish A. Kulkarni
- Department of Chemical Engineering University of Massachusetts Amherst MA 01003 USA
- Center for Bioactive Delivery Institute for Applied Life Sciences University of Massachusetts Amherst MA 01003 USA
| |
Collapse
|
45
|
Zhao M, Zhu T, Chen J, Cui Y, Zhang X, Lee RJ, Sun F, Li Y, Teng L. PLGA/PCADK composite microspheres containing hyaluronic acid-chitosan siRNA nanoparticles: A rational design for rheumatoid arthritis therapy. Int J Pharm 2021; 596:120204. [PMID: 33493604 DOI: 10.1016/j.ijpharm.2021.120204] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 12/14/2020] [Accepted: 01/01/2021] [Indexed: 12/27/2022]
Abstract
Myeloid cell leukemia-1 (Mcl-1), a member of the Bcl-2 anti-apoptotic family, is overexpressed in the synovial macrophages of patients with rheumatoid arthritis (RA). Small interfering RNA (siRNA) Mcl-1 can induce macrophage apoptosis in the joints and is a potential therapeutic target of RA. Nevertheless, the application of siRNA is limited owing to its instability and susceptibility to degradation in vivo. To address these shortcomings, we developed composite microspheres (MPs) loaded with hyaluronic acid (HA)-chitosan (CS) nanoparticles (NPs). First, we synthesized HA-CS/siRNA NPs (HCNPs) using ionotropic gelation process. Then, HCNPs, as an internal aqueous phase, were loaded into poly (D, L-lactide-co-glycolide) (PLGA) and poly (cyclohexane-1,4-diyl acetone dimethylene ketal) (PCADK) MPs using the double emulsion method. The NPs-in-MPs (NiMPs) composite system provided sustained release of NPs, protected siRNA against nuclease degradation in the serum, and could readily cross the cellular membrane. In addition, we evaluated the advantages of NiMPs in an adjuvant-induced arthritis rat model. Our experimental results demonstrate that NiMPs have greater pharmacodynamic effects than common MPs. Meanwhile, compared with HCNPs, NiMPs reduced the frequency of drug administration. Therefore, NiMPs are a promising and novel siRNA delivery vehicle for RA therapy.
Collapse
Affiliation(s)
- Menghui Zhao
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Tianyu Zhu
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Jicong Chen
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Yaxin Cui
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Xueyan Zhang
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Robert J Lee
- School of Life Sciences, Jilin University, Changchun, Jilin, China; College of Pharmacy, the Ohio State University, Columbus, OH, USA
| | - Fengying Sun
- School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Youxin Li
- School of Life Sciences, Jilin University, Changchun, Jilin, China.
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
46
|
Ashrafizadeh M, Delfi M, Hashemi F, Zabolian A, Saleki H, Bagherian M, Azami N, Farahani MV, Sharifzadeh SO, Hamzehlou S, Hushmandi K, Makvandi P, Zarrabi A, Hamblin MR, Varma RS. Biomedical application of chitosan-based nanoscale delivery systems: Potential usefulness in siRNA delivery for cancer therapy. Carbohydr Polym 2021; 260:117809. [PMID: 33712155 DOI: 10.1016/j.carbpol.2021.117809] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022]
Abstract
Gene therapy is an emerging and promising strategy in cancer therapy where small interfering RNA (siRNA) system has been deployed for down-regulation of targeted gene and subsequent inhibition in cancer progression; some issues with siRNA, however, linger namely, its off-targeting property and degradation by enzymes. Nanoparticles can be applied for the encapsulation of siRNA thus enhancing its efficacy in gene silencing where chitosan (CS), a linear alkaline polysaccharide derived from chitin, with superb properties such as biodegradability, biocompatibility, stability and solubility, can play a vital role. Herein, the potential of CS nanoparticles has been discussed for the delivery of siRNA in cancer therapy; proliferation, metastasis and chemoresistance are suppressed by siRNA-loaded CS nanoparticles, especially the usage of pH-sensitive CS nanoparticles. CS nanoparticles can provide a platform for the co-delivery of siRNA and anti-tumor agents with their enhanced stability via chemical modifications. As pre-clinical experiments are in agreement with potential of CS-based nanoparticles for siRNA delivery, and these carriers possess biocompatibiliy and are safe, further studies can focus on evaluating their utilization in cancer patients.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey; Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey
| | - Masoud Delfi
- Department of Chemical Sciences, University of Naples "Federico II", Complesso Universitario Monte S. Angelo, Via Cintia, 80126 Naples, Italy
| | - Farid Hashemi
- PhD Student of Pharmacology, Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hossein Saleki
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Morteza Bagherian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negar Azami
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Seyed Omid Sharifzadeh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Soodeh Hamzehlou
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Pooyan Makvandi
- Centre for Materials Interface, Istituto Italiano di Tecnologia, Pontedera 56025, Pisa, Italy
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956 Istanbul, Turkey.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
| | - Rajender S Varma
- Regional Center of Advanced Technologies and Materials, Palacky University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic.
| |
Collapse
|
47
|
Combined inhibition of CD73 and ZEB1 by Arg-Gly-Asp (RGD)-targeted nanoparticles inhibits tumor growth. Colloids Surf B Biointerfaces 2021; 197:111421. [DOI: 10.1016/j.colsurfb.2020.111421] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/09/2020] [Accepted: 10/11/2020] [Indexed: 12/15/2022]
|
48
|
Bastaki S, Aravindhan S, Ahmadpour Saheb N, Afsari Kashani M, Evgenievich Dorofeev A, Karoon Kiani F, Jahandideh H, Beigi Dargani F, Aksoun M, Nikkhoo A, Masjedi A, Mahmoodpoor A, Ahmadi M, Dolati S, Namvar Aghdash S, Jadidi-Niaragh F. Codelivery of STAT3 and PD-L1 siRNA by hyaluronate-TAT trimethyl/thiolated chitosan nanoparticles suppresses cancer progression in tumor-bearing mice. Life Sci 2020; 266:118847. [PMID: 33309720 DOI: 10.1016/j.lfs.2020.118847] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/20/2020] [Accepted: 11/28/2020] [Indexed: 12/14/2022]
Abstract
Immunotherapy methods using potential tumor microenvironment modulators have elicited durable therapeutic responses in cancer treatment. Immune checkpoint molecule programmed cell death-ligand 1 (PD-L1) and oncogenic transcription factor STAT3 (signal transducer and activator of transcription-3) assigned as inhibitory targets of our study and particular delivery system designed to deliver small interfering RNAs (siRNAs) to silence the targeted genes. Generated trimethyl chitosan (TMC) and thiolated chitosan (TC) nanoparticles (NPs) conjugated with HIV-1-derived TAT peptide and HA (hyaluronic acid) exhibited eligible physicochemical characteristics, notable siRNA encapsulation, serum stability, non-toxicity, controlled siRNA release, and extensive cellular uptake by cancer cells. Dual inhibition with STAT3/PD-L1 siRNA-loaded HA-TAT-TMC-TC NPs led to promising results, including significant downregulation of PD-L1 and STAT3 genes, striking suppressive effects on proliferation, migration, and angiogenesis of breast and melanoma cancer cell lines, and restrained tumor growth in vivo. These findings infer the capability of HA-TAT-TMC-TC NPs containing STAT3/PD-L1 siRNAs as a novel tumor-suppressive candidate in cancer treatment.
Collapse
Affiliation(s)
- Shima Bastaki
- Immunology research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Surendar Aravindhan
- Department of Electronics and Communication Engineering, AL-AMEEN ENGINEERING COLLEGE (Autonomous), Erode, Tamilnadu 638104, India.
| | | | | | | | - Fariba Karoon Kiani
- Immunology research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hediyeh Jahandideh
- Immunology research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohsen Aksoun
- Immunology research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Nikkhoo
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Masjedi
- Immunology research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, School of Medicine, Imam Reza Medical Research & Training Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Simin Namvar Aghdash
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
49
|
Ghasemi-Chaleshtari M, Kiaie SH, Irandoust M, Karami H, Nabi Afjadi M, Ghani S, Aghaei Vanda N, Ghaderi Sede MJ, Ahmadi A, Masjedi A, Hassannia H, Atyabi F, Hojjat-Farsangi M, Namdar A, Ghalamfarsa G, Jadidi-Niaragh F. Concomitant blockade of A2AR and CTLA-4 by siRNA-loaded polyethylene glycol-chitosan-alginate nanoparticles synergistically enhances antitumor T-cell responses. J Cell Physiol 2020; 235:10068-10080. [PMID: 32488862 DOI: 10.1002/jcp.29822] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/17/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022]
Abstract
Inhibitory immune checkpoint (ICP) molecules are important immunosuppressive factors in a tumor microenvironment (TME). They can robustly suppress T-cell-mediated antitumor immune responses leading to cancer progression. Among the checkpoint molecules, cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) is one of the critical inhibitors of anticancer T-cell responses. Besides, the expression of adenosine receptor (A2AR) on tumor-infiltrating T cells potently reduces their function. We hypothesized that concomitant silencing of these molecules in T cells might lead to enhanced antitumor responses. To examine this assumption, we purified T cells from the tumor, spleen, and local lymph nodes of CT26 colon cancer-bearing mice and suppressed the expression of A2AR and CTLA-4 using the small interfering RNA (siRNA)-loaded polyethylene glycol-chitosan-alginate (PCA) nanoparticles. The appropriate physicochemical properties of the produced nanoparticles (NPs; size of 72 nm, polydispersive index [PDI] < 0.2, and zeta potential of 11 mV) resulted in their high efficiency in transfection and suppression of target gene expression. Following the silencing of checkpoint molecules, various T-cell functions, including proliferation, apoptosis, cytokine secretion, differentiation, and cytotoxicity were analyzed, ex vivo. The results showed that the generated nanoparticles had optimal physicochemical characteristics and significantly suppressed the expression of target molecules in T cells. Moreover, a concomitant blockade of A2AR and CTLA-4 in T cells could synergistically enhance antitumor responses through the downregulation of PKA, SHP2, and PP2Aα signaling pathways. Therefore, this combination therapy can be considered as a novel promising anticancer therapeutic strategy, which should be further investigated in subsequent studies.
Collapse
Affiliation(s)
- Mitra Ghasemi-Chaleshtari
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Mazandaran, Iran
| | - Seyed Hossein Kiaie
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahzad Irandoust
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadis Karami
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, University of Tarbiat Modares, Tehran, Iran
| | - Sepideh Ghani
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasimeh Aghaei Vanda
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Javad Ghaderi Sede
- Department of Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Armin Ahmadi
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, Alabama
| | - Ali Masjedi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Hassannia
- Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemeh Atyabi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Afshin Namdar
- Department of Oncology, Cross Cancer Institute, The University of Alberta, Edmonton, Alberta, Canada
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
50
|
Rostami N, Nikkhoo A, Khazaei-Poul Y, Farhadi S, Sadat Haeri M, Moghadaszadeh Ardebili S, Aghaei Vanda N, Atyabi F, Namdar A, Baghaei M, Haghnavaz N, Kazemi T, Yousefi M, Ghalamfarsa G, Sabz G, Jadidi-Niaragh F. Coinhibition of S1PR1 and GP130 by siRNA-loaded alginate-conjugated trimethyl chitosan nanoparticles robustly blocks development of cancer cells. J Cell Physiol 2020; 235:9702-9717. [PMID: 32424937 DOI: 10.1002/jcp.29781] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023]
Abstract
There is an interconnected network between S1P/sphingosine-1-phosphate receptor 1 (S1PR1), IL-6/glycoprotein 130 (GP130), and signal transducer and activator of transcription 3 (STAT3) signaling pathways in the tumor microenvironment, which leads to cancer progression. S1P/S1PR1 and IL-6/GP130 signaling pathways phosphorylate and activate STAT3, and it then induces the expression of S1PR1 and interleukin-6 (IL-6) in a positive feedback loop leading to cancer progression. We hypothesized that blockade of this amplification loop can suppress the growth and development of cancer cells. Therefore, we silenced STAT3 upstream molecules including the S1PR1 and GP130 molecules in cancer cells using small interfering RNA (siRNA)-loaded alginate-conjugated trimethyl chitosan (ATMC) nanoparticles (NPs). The generated NPs had competent properties including the appropriate size, zeta potential, polydispersity index, morphology, high uptake of siRNA, high rate of capacity, high stability, and low toxicity. We evaluated the effects of siRNA loaded ATMC NPs on tumor hallmarks of three murine-derived cancer cell lines, including 4T1 (breast cancer), B16-F10 (melanoma), and CT26 (colon cancer). The results confirmed the tumor-suppressive effects of combinational targeting of S1PR1 and GP130. Moreover, combination therapy could potently suppress tumor growth as assessed by the chick chorioallantoic membrane assay. In this study, we targeted this positive feedback loop for the first time and applied this novel combination therapy, which provides a promising approach for cancer treatment. The development of a potent nanocarrier system with ATMC for this combination was also another aspect of this study, which should be further investigated in cancer animal models in further studies.
Collapse
Affiliation(s)
- Narges Rostami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Nikkhoo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yalda Khazaei-Poul
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shohreh Farhadi
- Student Research Committee, Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Melika Sadat Haeri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Fatemeh Atyabi
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Afshin Namdar
- Department of Oncology, Cross Cancer Institute, The University of Alberta, Edmonton, Alberta, Canada
| | - Masoumeh Baghaei
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Navideh Haghnavaz
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghasem Ghalamfarsa
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Gholamabas Sabz
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|