1
|
Wu C, Gao M, Xiao W, Huang X, Yang X, Wu Z, Yu X, Mo B, Du Z, Shang Z, Liu J, Shi C, Li R, Luo S, Wang W. Light-activatable manganese carbonate nanocubes elicit robust immunotherapy by amplifying endoplasmic reticulum stress-meditated pyroptotic cell death. J Exp Clin Cancer Res 2025; 44:147. [PMID: 40380194 DOI: 10.1186/s13046-025-03408-5] [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: 02/09/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025] Open
Abstract
Although tumor immunotherapy has emerged as a promising treatment modality, it faces significant challenges stemming from the immunosuppressive characteristics of the tumor microenvironment (TME), the low immunogenicity of tumors, and the poor specificity of immunoactivation. These factors can hinder the efficacy of immunotherapeutic approaches and lead to immune-related adverse events. This study reports a multifunctional nanocube (Mn-ER-Cy) that integrates Mn carbonate (MnCO3) and a photosensitizer (ER-Cy) by targeting tumor-cell endoplasmic reticulum (ER). The results demonstrate that Mn-ER-Cy preferentially accumulates in tumor tissues and is retained within ER organelles, facilitating photothermal therapy (PTT) and photodynamic therapy (PDT) upon exposure to 808 nm light irradiation. Triggered by acidic TME and light irradiation, MnCO3 is rapidly degraded to Mn2+, which in turn promotes the generation of reactive oxygen species through the Mn2+-mimic Fenton reaction, enabling chemical dynamics therapy (CDT). Triple-modal synergistic therapy simultaneously happens in ER to induce excessive ER stress, which subsequently amplify highly immunogenic pyroptotic cell death through activating NLRP3 inflammasome, caspase-1, and gasdermin D (GSDMD) pathway. Meanwhile, the decomposition of MnCO3 consumes H+ and contributes to an increased intracellular pH by regulating lactic acid levels, thereby counteracting the immunosuppressive acidic TME. Furthermore, Mn-ER-Cy serves as an inherent dual-modality imaging contrast agent for near-infrared fluorescence and photoacoustic imaging, facilitating imaging-guided precision therapy. These findings underscore the potential of Mn-ER-Cy to substantially enhance the efficacy and specificity of tumor immunotherapy, portraying a bright prospect to improve the clinical outcomes of patients with cancer.
Collapse
Affiliation(s)
- Chuan Wu
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Mingquan Gao
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Weidong Xiao
- Department of Pharmacy, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Xie Huang
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xinrui Yang
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zifei Wu
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xudong Yu
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Banghui Mo
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zaizhi Du
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ziqian Shang
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jing Liu
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Can Shi
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Rong Li
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Shenglin Luo
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Weidong Wang
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China.
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, 610041, China.
| |
Collapse
|
2
|
Ma X, Cong R, Cui X, Tang Y, Ren J, Hou J, Liu B, Zhao J, Li P, Li L, Zhang H, Tu J, Jiang L. Dendritic lipopeptide-based transdermal siRNA delivery systems for effective non-invasive therapy in psoriasis. J Control Release 2025; 381:113581. [PMID: 40020928 DOI: 10.1016/j.jconrel.2025.113581] [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: 10/17/2024] [Revised: 02/02/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Psoriasis is a prevalent chronic inflammatory skin disease characterized by immune cell activation and aberrant keratinocyte proliferation. Signal transducer and activator of transcription 3 (STAT3) plays a key role in the pathology of psoriasis, positioning it as a promising target for therapeutic strategies. However, current STAT3 inhibitors often lack specificity, leading to adverse effects. Here, we present the development of dendritic lipopeptides (DLPs) designed to facilitate the transdermal delivery of small interfering RNA (siRNA) to specifically inhibit STAT3 expression in psoriatic lesions. The dendritic architecture and peptide composition of DLP are crucial for interaction with keratin of stratum corneum and gene transfection efficiency, while the lipid chain selection aims to increase lipophilicity and enhance interactions with cellular membranes. We conducted extensive in vitro and in vivo investigations to assess the therapeutic efficacy of these DLPs for siRNA delivery in psoriasis treatment. The results demonstrated that our DLPs effectively penetrated the skin barrier, delivered siRNA to target cells, and significantly reduced STAT3 expression, regulated immune cell imbalance, leading to a marked improvement in psoriasis symptoms. In conclusion, our study presents a promising non-invasive approach to psoriasis treatment by focusing on the targeted suppression of STAT3 expression. The dendritic lipopeptides offer a safe and effective platform of siRNA delivery, potentially revolutionizing the management of psoriasis and other chronic inflammatory dermatological conditions.
Collapse
Affiliation(s)
- Xiaolei Ma
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Rui Cong
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients and Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Xin Cui
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients and Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yiwei Tang
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients and Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Jie Ren
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients and Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Jinfan Hou
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients and Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Boyu Liu
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients and Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Jiehui Zhao
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients and Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Pengwei Li
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients and Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Lei Li
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients and Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Heyang Zhang
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC Leiden, Netherlands
| | - Jiasheng Tu
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients and Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| | - Lei Jiang
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients and Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| |
Collapse
|
3
|
Markovic MD, Panic VV, Pjanovic RV. Polymeric Nanosystems: A Breakthrough Approach to Treating Inflammation and Inflammation Related Diseases. Biopolymers 2025; 116:e70012. [PMID: 40104970 DOI: 10.1002/bip.70012] [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: 09/25/2024] [Revised: 02/17/2025] [Accepted: 03/02/2025] [Indexed: 03/20/2025]
Abstract
Inflammation processes can cause mild to severe damage in the human body and can lead to a large number of inflammation-related diseases (IRD) such as cancer, neural, vascular, and pulmonary diseases. Limitations of anti-inflammatory drugs (AID) application are reflected in high therapeutic doses, toxicity, low bioavailability and solubility, side effects, etc. Polymeric nanosystems (PS) have been recognized as a safe and effective technology that is able to overcome these limitations by AID encapsulation and is able to answer to the specific demands of the IRD treatment. PS are attracting great attention due to their versatility, biocompatibility, low toxicity, fine-tuned properties, functionality, and ability for precise delivery of anti-inflammatory drugs to the targeted sites in the human body. This article offers an overview of three classes of polymeric nanosystems: a) dendrimers, b) polymeric micelles and polymeric nanoparticles, and c) polymeric filomicelles, as well as their properties, preparation, and application in IRD treatment. In the future, the number of PS formulations in clinical practice will certainly increase.
Collapse
Affiliation(s)
- Maja D Markovic
- Innovation Center of Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Vesna V Panic
- Innovation Center of Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Rada V Pjanovic
- Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
4
|
Miao Y, Wang K, Liu X, Wang X, Hu Y, Yuan Z, Deng D. Multifunctional biomimetic liposomal nucleic acid scavengers inhibit the growth and metastasis of breast cancer. Biomater Sci 2025; 13:2475-2488. [PMID: 40152107 DOI: 10.1039/d4bm01721h] [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: 03/29/2025]
Abstract
Chemotherapy and surgery, though effective in cancer treatment, trigger the release of nucleic acid-containing pro-inflammatory compounds from damaged tumor cells, known as nucleic acid-associated damage-associated molecular patterns (NA-DAMPs). This inflammation promotes tumor metastasis, and currently, no effective treatment exists for this treatment-induced inflammation and subsequent tumor metastasis. To address this challenge, we developed a biomimetic liposome complex (Lipo-Rh2) incorporating a hybrid structure of liposomes and dendritic polymers, mimicking cell membrane morphology. Lipo-Rh2 leverages the multivalent surface properties of dendritic polymers to clear cell-free nucleic acids while serving as both a structural stabilizer and targeting ligand via embedded ginsenoside Rh2. Experimental data show that Lipo-Rh2 effectively reduces free nucleic acids in mouse serum through charge interactions, downregulates Toll-like receptor expression, decreases inflammatory cytokine secretion, and inhibits both primary tumor growth and metastasis. Compared to the current nucleic acid scavenger PAMAM-G3, Lipo-Rh2 demonstrates stronger antitumor effects, lower toxicity, and enhanced targeting capabilities. This biomimetic liposome-based nucleic acid scavenger represents a novel approach to nucleic acid clearance, expanding the framework for designing effective therapeutic agents.
Collapse
Affiliation(s)
- Yuhang Miao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China.
| | - Kaizhen Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China.
| | - Xin Liu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Xin Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China.
| | - Yanwei Hu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Zhenwei Yuan
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China.
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Dawei Deng
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China.
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
5
|
Yang R, Xu K, Li H, Feng Y, Xiang G, Zhou X, Zhang C. Laminarin-mediated oral delivery of miRNA-223 for targeted macrophage polarization in inflammatory bowel disease. Int J Biol Macromol 2025; 311:143052. [PMID: 40294678 DOI: 10.1016/j.ijbiomac.2025.143052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/30/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025]
Abstract
Intestinal inflammatory disorders, such as ulcerative colitis, are driven by excessive pro-inflammatory M1 macrophages, leading to tissue damage and chronic inflammation. MicroRNA-223 (miRNA-223) presents a promising therapeutic approach by shifting macrophage polarization from the M1 to the anti-inflammatory M2 phenotype, thereby reducing inflammation. However, oral delivery of miRNA-223 is hindered by its instability in the harsh gastrointestinal (GI) environment and the challenge of targeting colonic macrophages. To overcome these obstacles, we developed an oral nanogene delivery system using laminarin, a natural β-1,3 glucan polysaccharide derived from kelp, as the primary delivery unit. miRNA-223 is encapsulated within peptide dendrimers, which are then coated with laminarin to enhance stability and enable targeted delivery via dectin-1 receptors on macrophages. This laminarin-mediated formulation protects miRNA-223 from degradation in the GI tract and facilitates its selective uptake by inflamed colonic macrophages. In vitro and in vivo studies demonstrated that the laminarin-coated nanogene complex preserves miRNA-223 integrity, targets inflamed colonic regions, and promotes M2 polarization, resulting in reduced inflammation and improved outcomes in a colitis mouse model. This work highlights the potential of natural laminarin-based nanocarriers for effective oral miRNA delivery, offering a novel strategy for treating inflammatory bowel diseases through precise modulation of macrophage phenotypes.
Collapse
Affiliation(s)
- Ran Yang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The First People's Hospital of Jintang County, Sichuan Province, Chengdu 610400, China
| | - Kangkang Xu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Huihui Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Yuantao Feng
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Gang Xiang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Xing Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Rehabilitation School, Kunming Medical University, Kunming 650500, China.
| | - Chengyuan Zhang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Rehabilitation School, Kunming Medical University, Kunming 650500, China.
| |
Collapse
|
6
|
Saha S, Tandon R, Sanku J, Kumari A, Shukla R, Srivastava N. siRNA-based Therapeutics in Hormone-driven Cancers: Advancements and benefits over conventional treatments. Int J Pharm 2025; 674:125463. [PMID: 40081431 DOI: 10.1016/j.ijpharm.2025.125463] [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: 11/19/2024] [Revised: 02/10/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Hormone-related cancers, also known as hormone-sensitive or hormone-dependent cancers, rely on hormones such as estrogen, testosterone, and progesterone for growth. These malignancies, including breast, pituitary, thyroid, ovarian, uterine, cervical, and prostate cancers, often exhibit accelerated progression in response to hormonal signaling. Small interfering RNA (siRNA) has emerged as a groundbreaking gene suppression therapy since the FDA approval of its first product in 2018. With over 200 ongoing clinical trials, siRNA is being actively explored as a targeted treatment for hormone-related cancers. Its ability to silence specific oncogenes offers significant advantages over conventional therapies, which are often associated with toxicity, resistance, and non-specific targeting. However, challenges in siRNA delivery remain a major barrier to its clinical translation, limiting its ability to reach target cells effectively. This review evaluates the potential of siRNA in hormone-related cancers, addressing the shortcomings of traditional treatments while examining novel strategies to enhance siRNA delivery and overcome tumor microenvironment obstacles. Notably, no existing literature comprehensively consolidates siRNA-based therapies for these cancers, emphasizing the importance of this manuscript in bridging current knowledge gaps and advancing the translational application of siRNA therapeutics.
Collapse
Affiliation(s)
- Sayani Saha
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Reetika Tandon
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Jhansi Sanku
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Anchala Kumari
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India.
| |
Collapse
|
7
|
Ma M, Li X, Zhong M, Li X, Yu J, Wang Z, Lv Q, Li X, He Z, Liu H, Wang Y. Galloylated Toll-Like Receptor 7/8 Agonist Nanovaccine for Enhanced Tumor Antigen Delivery in Personalized Immunotherapy. ACS NANO 2025; 19:11900-11912. [PMID: 40102033 DOI: 10.1021/acsnano.4c15442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Cancer vaccines, a critical technology in cancer immunotherapy, have shown great therapeutic potential. However, traditional vaccines based on tumor cell lysates (TCLs) have shown disappointing results in early clinical trials due to low immunogenicity, in vivo instability, and the inability to codeliver with adjuvants. To address these issues, we developed a nanoparticle vaccine, R848-GA@TCLs, by modifying the toll-like receptor 7/8 (TLR7/8) agonist R848 with gallic acid. This nanovaccine leverages the "capturing" ability of the galloyl moiety to coload TCLs and R848, forming stable nanoparticles. R848-GA@TCLs efficiently target lymph nodes, increasing TCL accumulation 10-fold, and enable the synchronized release of antigens and adjuvants within dendritic cells (DCs). Our results show that R848-GA@TCLs increase with respect to the cross-presentation of tumor antigens, promote the production of pro-inflammatory cytokines, and activate DCs, leading to a significant increase in effector T cells, natural killer (NK) cells, and M1 macrophages. This strong immune response resulted in potent antitumor effects, with R848-GA@TCLs demonstrating efficacy in multiple tumor models by significantly inhibiting tumor growth and metastasis. In conclusion, R848-GA@TCLs represent a personalized cancer vaccine capable of codelivering TCLs and adjuvants, eliciting robust antitumor immune responses, and hold great potential for clinical applications.
Collapse
Affiliation(s)
- Mengyao Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Ximu Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Mingyuan Zhong
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Xuejing Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jiang Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zhaomeng Wang
- Department of Oncology, Innovative Cancer Drug Research and Engineering Center of Liaoning Province, Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Qingzhi Lv
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Xin Li
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| |
Collapse
|
8
|
Tang C, Zhang Y, Li B, Fan X, Wang Z, Su R, Qi W, Wang Y. Modular Design of Lipopeptide-Based Organ-Specific Targeting (POST) Lipid Nanoparticles for Highly Efficient RNA Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415643. [PMID: 39924757 DOI: 10.1002/adma.202415643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/17/2025] [Indexed: 02/11/2025]
Abstract
Lipid nanoparticles (LNPs) with highly efficient and specific extrahepatic targeting abilities are promising in gene delivery, and the lipopeptides (LPs) with excellent designability and functionality are expected to empower the construction of functional LNPs. This study aims to develop highly efficient ionizable components that accurately match different targeting lipid systems through the modular design of LPs. Based on this, a lipopeptide-based organ-specific targeting (POST) LNP screening strategy is constructed, in which lysine-histidine-based lipopeptides (KH-LPs) are designed as highly efficient ionizable components. The optimal KH-LP LNP screened in vitro shows excellent siRNA/mRNA transfecting ability in various hard-to-transfect cell lines. Compared to the classic LNPs, the POST LNPs screened in vivo achieve even higher (or at least comparable) efficiency and specificity in delivering mRNA and siRNA to the lung, liver, and spleen, respectively. The structure-activity relationship (SAR) proves that the modular regulation of LP structures can accurately provide the optimal ionizable components for different targeting lipid systems, demonstrating the potential of this strategy in developing efficient and selective targeting systems, which is expected to open up more possibilities for gene therapy.
Collapse
Affiliation(s)
- Chuanmei Tang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Yexi Zhang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Bowen Li
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Xiangwei Fan
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Zixuan Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Rongxin Su
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Wei Qi
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Yuefei Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin, 300072, P. R. China
| |
Collapse
|
9
|
Yang Q, Hu Z, Jiang H, Wang J, Han H, Shi W, Qian H. Recent advances, strategies, and future perspectives of peptide-based drugs in clinical applications. Chin J Nat Med 2025; 23:31-42. [PMID: 39855829 DOI: 10.1016/s1875-5364(25)60800-4] [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: 07/07/2024] [Revised: 08/15/2024] [Accepted: 09/01/2024] [Indexed: 01/27/2025]
Abstract
Peptide-based therapies have attracted considerable interest in the treatment of cancer, diabetes, bacterial infections, and neurodegenerative diseases due to their promising therapeutic properties and enhanced safety profiles. This review provides a comprehensive overview of the major trends in peptide drug discovery and development, emphasizing preclinical strategies aimed at improving peptide stability, specificity, and pharmacokinetic properties. It assesses the current applications and challenges of peptide-based drugs in these diseases, illustrating the pharmaceutical areas where peptide-based drugs demonstrate significant potential. Furthermore, this review analyzes the obstacles that must be overcome in the future, aiming to provide valuable insights and references for the continued advancement of peptide-based drugs.
Collapse
Affiliation(s)
- Qimeng Yang
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Zhipeng Hu
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hongyu Jiang
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jialing Wang
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Han Han
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Wei Shi
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Hai Qian
- Centre of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
10
|
Tupally KR, Seal P, Pandey P, Lohman R, Smith S, Ouyang D, Parekh H. Integration of Dendrimer‐Based Delivery Technologies with Computational Pharmaceutics and Their Potential in the Era of Nanomedicine. EXPLORING COMPUTATIONAL PHARMACEUTICS ‐ AI AND MODELING IN PHARMA 4.0 2024:328-378. [DOI: 10.1002/9781119987260.ch10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
11
|
Warthen JL, Lueckheide MJ. Peptides as Targeting Agents and Therapeutics: A Brief Overview. Biomacromolecules 2024; 25:6923-6935. [PMID: 39445576 DOI: 10.1021/acs.biomac.4c00518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
The controllability and specificity of peptides make them ideal for targeting therapeutic delivery systems and as therapeutic agents that interfere with the essential functions of pathogens and tumors. Peptides can also mimic natural protein structures or parts thereof, agonize receptors, and be conjugated to other molecules that will self-assemble. In this short Review, we discuss research from the last ten years into peptide use in three arenas: the treatment of cancer, the treatment of pathogens, and the targeting of specific organs and organelles. These studies demonstrate the successful application of targeting and therapeutic peptides in vitro and in vivo and show the promising range of applications peptides can have going forward.
Collapse
Affiliation(s)
- Jalissa L Warthen
- Adelphi University, 1 South Avenue, Garden City, New York 11530, United States
| | | |
Collapse
|
12
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [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: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
13
|
Chen P, Wang Z, Wang X, Gong J, Sheng J, Pan Y, Zhu D, Liu X. ROS-Responsive Ferrocenyl Amphiphilic PAMAM Dendrimers for On-Demand Delivery of siRNA Therapeutics to Cancer Cells. Pharmaceutics 2024; 16:936. [PMID: 39065632 PMCID: PMC11280363 DOI: 10.3390/pharmaceutics16070936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/26/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Small interfering RNA (siRNA) therapeutics, characterized by high specificity, potency, and durability, hold great promise in the treatment of cancer and other diseases. However, the clinic implementation of siRNA therapeutics critically depends on the safe and on-demand delivery of siRNA to the target cells. Here, we reported a family of ferrocenyl amphiphilic dendrimers (Fc-AmDs) for on-demand delivery of siRNA in response to the high ROS content in cancer cells. These dendrimers bear ROS-sensitive ferrocene moieties in the hydrophobic components and positively chargeable poly(amidoamine) dendrons as the hydrophilic entities, possessing favorable safety profiles and ROS responsive properties. One of these ferrocenyl amphiphilic dendrimers, Fc-C8-AmD 8A, outperforms in siRNA delivery, benefiting from its optimal balance of hydrophobicity and hydrophilicity. Its ROS feature facilitates specific and efficient disassembly of its complex with siRNA in ROS-rich cancer cells for effective siRNA delivery and gene silencing. Moreover, Fc-C8-AmD 8A also integrates the features and beneficial properties of both lipid and dendrimer vectors. Therefore, it represents a novel on-demand delivery system for cancer cell-specific siRNA delivery. This work opens new perspectives for designing self-assembly nanosystems for on-demand drug delivery.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dandan Zhu
- Center of Advanced Pharmaceuticals and Biomaterials, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; (P.C.); (Z.W.); (X.W.); (J.G.); (J.S.); (Y.P.)
| | - Xiaoxuan Liu
- Center of Advanced Pharmaceuticals and Biomaterials, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; (P.C.); (Z.W.); (X.W.); (J.G.); (J.S.); (Y.P.)
| |
Collapse
|
14
|
Sztandera K, Rodríguez-García JL, Ceña V. In Vivo Applications of Dendrimers: A Step toward the Future of Nanoparticle-Mediated Therapeutics. Pharmaceutics 2024; 16:439. [PMID: 38675101 PMCID: PMC11053723 DOI: 10.3390/pharmaceutics16040439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/17/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Over the last few years, the development of nanotechnology has allowed for the synthesis of many different nanostructures with controlled sizes, shapes, and chemical properties, with dendrimers being the best-characterized of them. In this review, we present a succinct view of the structure and the synthetic procedures used for dendrimer synthesis, as well as the cellular uptake mechanisms used by these nanoparticles to gain access to the cell. In addition, the manuscript reviews the reported in vivo applications of dendrimers as drug carriers for drugs used in the treatment of cancer, neurodegenerative diseases, infections, and ocular diseases. The dendrimer-based formulations that have reached different phases of clinical trials, including safety and pharmacokinetic studies, or as delivery agents for therapeutic compounds are also presented. The continuous development of nanotechnology which makes it possible to produce increasingly sophisticated and complex dendrimers indicates that this fascinating family of nanoparticles has a wide potential in the pharmaceutical industry, especially for applications in drug delivery systems, and that the number of dendrimer-based compounds entering clinical trials will markedly increase during the coming years.
Collapse
Affiliation(s)
- Krzysztof Sztandera
- Unidad Asociada Neurodeath, Instituto de Nanociencia Molecular, Universidad de Castilla-La Mancha, 02006 Albacete, Spain;
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Valentín Ceña
- Unidad Asociada Neurodeath, Instituto de Nanociencia Molecular, Universidad de Castilla-La Mancha, 02006 Albacete, Spain;
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
15
|
Dachani S, Kaleem M, Mujtaba MA, Mahajan N, Ali SA, Almutairy AF, Mahmood D, Anwer MK, Ali MD, Kumar S. A Comprehensive Review of Various Therapeutic Strategies for the Management of Skin Cancer. ACS OMEGA 2024; 9:10030-10048. [PMID: 38463249 PMCID: PMC10918819 DOI: 10.1021/acsomega.3c09780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 03/12/2024]
Abstract
Skin cancer (SC) poses a global threat to the healthcare system and is expected to increase significantly over the next two decades if not diagnosed at an early stage. Early diagnosis is crucial for successful treatment, as the disease becomes more challenging to cure as it progresses. However, identifying new drugs, achieving clinical success, and overcoming drug resistance remain significant challenges. To overcome these obstacles and provide effective treatment, it is crucial to understand the causes of skin cancer, how cells grow and divide, factors that affect cell growth, and how drug resistance occurs. In this review, we have explained various therapeutic approaches for SC treatment via ligands, targeted photosensitizers, natural and synthetic drugs for the treatment of SC, an epigenetic approach for management of melanoma, photodynamic therapy, and targeted therapy for BRAF-mutated melanoma. This article also provides a detailed summary of the various natural drugs that are effective in managing melanoma and reducing the occurrence of skin cancer at early stages and focuses on the current status and future prospects of various therapies available for the management of skin cancer.
Collapse
Affiliation(s)
- Sudharshan
Reddy Dachani
- Department
of Pharmacy Practice, College of Pharmacy, Shaqra University, Al-Dawadmi Campus, Al-Dawadmi 11961, Saudi Arabia
| | - Mohammed Kaleem
- Department
of Pharmacology, Babasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440037, Maharashtra, India
| | - Md. Ali Mujtaba
- Department
of Pharmaceutics, Faculty of Pharmacy, Northern
Border University, Arar 91911, Saudi Arabia
| | - Nilesh Mahajan
- Department
of Pharmaceutics, Dabasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440037, Maharashtra, India
| | - Sayyed A. Ali
- Department
of Pharmaceutics, Dabasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440037, Maharashtra, India
| | - Ali F Almutairy
- Department
of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Danish Mahmood
- Department
of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Md. Khalid Anwer
- Department
of Pharmaceutics, College of Pharmacy, Prince
Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Mohammad Daud Ali
- Department
of Pharmacy, Mohammed Al-Mana College for
Medical Sciences, Abdulrazaq Bin Hammam Street, Al Safa 34222, Dammam, Saudi Arabia
| | - Sanjay Kumar
- Department
of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Uttar Pradesh 201306, India
| |
Collapse
|
16
|
Wang J, Ye M, Zhu B. Peptide Self-Assembly Facilitating DNA Transfection and the Application in Inhibiting Cancer Cells. Molecules 2024; 29:932. [PMID: 38474444 DOI: 10.3390/molecules29050932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/03/2024] [Accepted: 02/11/2024] [Indexed: 03/14/2024] Open
Abstract
Non-viral vectors have been developing in gene delivery due to their safety and low immunogenicity. But their transfection effect is usually very low, thus limiting the application. Hence, we designed eight peptides (compounds 1-8). We compared their performances; compound 8 had the best transfection efficacy and biocompatibility. The transfection effect was similar with that of PEI, a most-widely-employed commercial transfection reagent. Atomic force microscope (AFM) images showed that the compound could self-assemble and the self-assembled peptide might encapsulate DNA. Based on these results, we further analyzed the inhibitory result in cancer cells and found that compound 8 could partially fight against Hela cells. Therefore, the compound is promising to pave the way for the development of more effective and less toxic transfection vectors.
Collapse
Affiliation(s)
- Jingyu Wang
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin 300070, China
| | - Min Ye
- College of Pharmacy, Southern Medical University, Guangzhou 510280, China
| | - Baokuan Zhu
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
17
|
Singh D, Irham LM, Singh A, Kurmi BD. Guanidinium-based Integrated Peptide Dendrimers: Pioneer Nanocarrier in Cancer Therapy. Protein Pept Lett 2024; 31:261-274. [PMID: 38629378 DOI: 10.2174/0109298665292042240325052536] [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: 12/12/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 08/13/2024]
Abstract
The landscape of cancer therapy has witnessed a paradigm shift with the emergence of innovative delivery systems, and Guanidinium-based Peptide Dendrimers have emerged as a vanguard in this transformative journey. With their unique molecular architecture and intrinsic biocompatibility, these dendrimers offer a promising avenue for the targeted delivery of therapeutic cargo in cancer treatment. This comprehensive review delves into the intricate world of Guanidinium- based Peptide Dendrimers, unraveling their structural intricacies, mechanisms of action, and advancements that have propelled them from laboratory curiosities to potential clinical champions. Exploiting the potent properties of guanidinium, these dendrimers exhibit unparalleled precision in encapsulating and transporting diverse cargo molecules, ranging from conventional chemotherapeutics to cutting-edge nucleic acids. The review navigates the depths of their design principles, investigating their prowess in traversing the complex terrain of cellular barriers for optimal cargo delivery. Moreover, it delves into emerging trends, such as personalized therapeutic approaches, multimodal imaging, and bioinformatics-driven design, highlighting their potential to redefine the future of cancer therapy. Crucially, the review addresses the pivotal concerns of biocompatibility and safety, examining cytotoxicity profiles, immune responses, and in vivo studies. It underscores the importance of aligning scientific marvels with the stringent demands of clinical applications. Through each section, the narrative underscores the promises and possibilities that Guanidinium-based Peptide Dendrimers hold and how they can potentially reshape the landscape of precision cancer therapy.
Collapse
Affiliation(s)
- Dilpreet Singh
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, 140413, India
- University Centre for Research and Development, Chandigarh University, Gharuan (140413), India
| | | | - Amrinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
18
|
Shchaslyvyi AY, Antonenko SV, Tesliuk MG, Telegeev GD. Current State of Human Gene Therapy: Approved Products and Vectors. Pharmaceuticals (Basel) 2023; 16:1416. [PMID: 37895887 PMCID: PMC10609992 DOI: 10.3390/ph16101416] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/13/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
In the realm of gene therapy, a pivotal moment arrived with Paul Berg's groundbreaking identification of the first recombinant DNA in 1972. This achievement set the stage for future breakthroughs. Conditions once considered undefeatable, like melanoma, pancreatic cancer, and a host of other ailments, are now being addressed at their root cause-the genetic level. Presently, the gene therapy landscape stands adorned with 22 approved in vivo and ex vivo products, including IMLYGIC, LUXTURNA, Zolgensma, Spinraza, Patisiran, and many more. In this comprehensive exploration, we delve into a rich assortment of 16 drugs, from siRNA, miRNA, and CRISPR/Cas9 to DNA aptamers and TRAIL/APO2L, as well as 46 carriers, from AAV, AdV, LNPs, and exosomes to naked mRNA, sonoporation, and magnetofection. The article also discusses the advantages and disadvantages of each product and vector type, as well as the current challenges faced in the practical use of gene therapy and its future potential.
Collapse
Affiliation(s)
- Aladdin Y. Shchaslyvyi
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, 150, Zabolotnogo Str., 03143 Kyiv, Ukraine; (S.V.A.); (M.G.T.); (G.D.T.)
| | | | | | | |
Collapse
|
19
|
Gu F, Huang X, Huang W, Zhao M, Zheng H, Wang Y, Chen R. The role of miRNAs in Behçet's disease. Front Immunol 2023; 14:1249826. [PMID: 37860009 PMCID: PMC10584330 DOI: 10.3389/fimmu.2023.1249826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/12/2023] [Indexed: 10/21/2023] Open
Abstract
The symptoms of Behçet's disease (BD), a multisystemic condition with autoimmune and inflammation as hallmarks, include arthritis, recurring oral and vaginal ulcers, skin rashes and lesions, and involvement of the nervous, gastrointestinal, and vascular systems. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), may be important regulators of inflammation and autoimmune disease. These ncRNAs are essential to the physiological and pathophysiological disease course, and miRNA in particular has received significant attention for its role and function in BD and its potential use as a diagnostic biomarker in recent years. Although promising as therapeutic targets, miRNAs must be studied further to fully comprehend how miRNAs in BD act biologically.
Collapse
Affiliation(s)
| | | | | | | | | | - Yuanyin Wang
- College and Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, China
| | - Ran Chen
- College and Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, China
| |
Collapse
|
20
|
Klipp A, Burger M, Leroux JC. Get out or die trying: Peptide- and protein-based endosomal escape of RNA therapeutics. Adv Drug Deliv Rev 2023; 200:115047. [PMID: 37536508 DOI: 10.1016/j.addr.2023.115047] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/28/2023] [Accepted: 08/01/2023] [Indexed: 08/05/2023]
Abstract
RNA therapeutics offer great potential to transform the biomedical landscape, encompassing the treatment of hereditary conditions and the development of better vaccines. However, the delivery of RNAs into the cell is hampered, among others, by poor endosomal escape. This major hurdle is often tackled using special lipids, polymers, or protein-based delivery vectors. In this review, we will focus on the most prominent peptide- and protein-based endosomal escape strategies with focus on RNA drugs. We discuss cell penetrating peptides, which are still incorporated into novel transfection systems today to promote endosomal escape. However, direct evidence for enhanced endosomal escape by the action of such peptides is missing and their transfection efficiency, even in permissive cell culture conditions, is rather low. Endosomal escape by the help of pore forming proteins or phospholipases, on the other hand, allowed to generate more efficient transfection systems. These are, however, often hampered by considerable toxicity and immunogenicity. We conclude that the perfect enhancer of endosomal escape has yet to be devised. To increase the chances of success, any new transfection system should be tested under relevant conditions and guided by assays that allow direct quantification of endosomal escape.
Collapse
Affiliation(s)
- Alexander Klipp
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland.
| | - Michael Burger
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland.
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Switzerland.
| |
Collapse
|
21
|
Sharma S, Mahajan SD, Chevli K, Schwartz SA, Aalinkeel R. Nanotherapeutic Approach to Delivery of Chemo- and Gene Therapy for Organ-Confined and Advanced Castration-Resistant Prostate Cancer. Crit Rev Ther Drug Carrier Syst 2023; 40:69-100. [PMID: 37075068 PMCID: PMC11007628 DOI: 10.1615/critrevtherdrugcarriersyst.2022043827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Treatments for late-stage prostate cancer (CaP) have not been very successful. Frequently, advanced CaP progresses to castration-resistant prostate cancer (CRPC), with 50#37;-70% of patients developing bone metastases. CaP with bone metastasis-associated clinical complications and treatment resistance presents major clinical challenges. Recent advances in the formulation of clinically applicable nanoparticles (NPs) have attracted attention in the fields of medicine and pharmacology with applications to cancer and infectious and neurological diseases. NPs have been rendered biocompatible, pose little to no toxicity to healthy cells and tissues, and are engineered to carry large therapeutic payloads, including chemo- and genetic therapies. Additionally, if required, targeting specificity can be achieved by chemically coupling aptamers, unique peptide ligands, or monoclonal antibodies to the surface of NPs. Encapsulating toxic drugs within NPs and delivering them specifically to their cellular targets overcomes the problem of systemic toxicity. Encapsulating highly labile genetic therapeutics such as RNA within NPs provides a protective environment for the payload during parenteral administration. The loading efficiencies of NPs have been maximized while the controlled their therapeutic cargos has been released. Theranostic ("treat and see") NPs have developed combining therapy with imaging capabilities to provide real-time, image-guided monitoring of the delivery of their therapeutic payloads. All of these NP accomplishments have been applied to the nanotherapy of late-stage CaP, offering a new opportunity for a previously dismal prognosis. This article gives an update on current developments in the use of nanotechnology for treating late-stage, castration-resistant CaP.
Collapse
Affiliation(s)
- Satish Sharma
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Supriya D. Mahajan
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Kent Chevli
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Stanley A. Schwartz
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Ravikumar Aalinkeel
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| |
Collapse
|
22
|
Subhan MA, Torchilin VP. Biopolymer-Based Nanosystems for siRNA Drug Delivery to Solid Tumors including Breast Cancer. Pharmaceutics 2023; 15:pharmaceutics15010153. [PMID: 36678782 PMCID: PMC9861964 DOI: 10.3390/pharmaceutics15010153] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Nanobiopolymers such as chitosan, gelatin, hyaluronic acid, polyglutamic acid, lipids, peptides, exosomes, etc., delivery systems have prospects to help overwhelmed physiological difficulties allied with the delivery of siRNA drugs to solid tumors, including breast cancer cells. Nanobiopolymers have favorable stimuli-responsive properties and therefore can be utilized to improve siRNA delivery platforms to undruggable MDR metastatic cancer cells. These biopolymeric siRNA drugs can shield drugs from pH degradation, extracellular trafficking, and nontargeted binding sites and are consequently suitable for drug internalization in a controlled-release fashion. In this review, the utilization of numerous biopolymeric compounds such as siRNA drug delivery systems for MDR solid tumors, including breast cancers, will be discussed.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh
- Correspondence: (M.A.S.); (V.P.T.)
| | - Vladimir P. Torchilin
- CPBN, Department of Pharmaceutical Sciences, North Eastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, North Eastern University, Boston, MA 02115, USA
- Correspondence: (M.A.S.); (V.P.T.)
| |
Collapse
|
23
|
A combinatorial delivery of survivin targeted siRNA using cancer selective nanoparticles for triple negative breast cancer therapy. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
24
|
Yadav DN, Ali MS, Thanekar AM, Pogu SV, Rengan AK. Recent Advancements in the Design of Nanodelivery Systems of siRNA for Cancer Therapy. Mol Pharm 2022; 19:4506-4526. [PMID: 36409653 DOI: 10.1021/acs.molpharmaceut.2c00811] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
RNA interference (RNAi) has increased the possibility of restoring RNA drug targets for cancer treatment. Small interfering RNA (siRNA) is a promising therapeutic RNAi tool that targets the defective gene by inhibiting its mRNA expression and stopping its translation. However, siRNAs have flaws like poor intracellular trafficking, RNase degradation, rapid kidney filtration, off-targeting, and toxicity, which limit their therapeutic efficiency. Nanocarriers (NCs) have been designed to overcome such flaws and increase antitumor activity. Combining siRNA and anticancer drugs can give synergistic effects in cancer cells, making them a significant gene-modification tool in cancer therapy. Our discussion of NCs-mediated siRNA delivery in this review includes their mechanism, limitations, and advantages in comparison with naked siRNA delivery. We will also discuss organic NCs (polymers and lipids) and inorganic NCs (quantum dots, carbon nanotubes, and gold) that have been reported for extensive delivery of therapeutic siRNA to tumor sites. Finally, we will conclude by discussing the studies based on organic and inorganic NCs-mediated siRNA drug delivery systems conducted in the years 2020 and 2021.
Collapse
Affiliation(s)
- Dokkari Nagalaxmi Yadav
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad, Kandi 502284, India
| | - Mohammad Sadik Ali
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad, Kandi 502284, India
| | | | - Sunil Venkanna Pogu
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad, Kandi 502284, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad, Kandi 502284, India
| |
Collapse
|
25
|
Ke J, Zhang J, Li J, Liu J, Guan S. Design of Cyclic Peptide-Based Nanospheres and the Delivery of siRNA. Int J Mol Sci 2022; 23:ijms232012071. [PMID: 36292932 PMCID: PMC9602810 DOI: 10.3390/ijms232012071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/29/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
In recent years, cyclic peptides have attracted much attention due to their chemical and enzymatic stability, low toxicity, and easy modification. In general, the self-assembled nanostructures of cyclic peptides tend to form nanotubes in a cyclic stacking manner through hydrogen bonding. However, studies exploring other assembly strategies are scarce. In this context, we proposed a new assembly strategy based on cyclic peptides with covalent self-assembly. Here, cyclic peptide-(DPDPDP) was rationally designed and used as a building block to construct new assemblies. With cyclo-(DP)3 as the structural unit and 2,2′-diamino-N-methyldiethylamine as the linker, positively charged nanospheres ((CP)6NS) based on cyclo-(DP)3 were successfully constructed by covalent self-assembly. We assessed their size and morphology by scanning electron microscopy (SEM), TEM, and DLS. (CP)6NS were found to have a strong positive charge, so they could bind to siRNA through electrostatic interactions. Confocal microscopy analysis and cell viability assays showed that (CP)6NS had high cellular internalization efficiency and low cytotoxicity. More importantly, real-time polymerase chain reaction (PCR) and flow cytometry analyses indicated that (CP)6NS-siRNA complexes potently inhibited gene expression and promoted tumor cell apoptosis. These results suggest that (CP)6NS may be a potential siRNA carrier for gene therapy.
Collapse
Affiliation(s)
- Junfeng Ke
- School of Life Sciences, Jilin University, Changchun 130012, China
- Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun 130012, China
| | - Jingli Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China
- Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun 130012, China
| | - Junyang Li
- School of Life Sciences, Jilin University, Changchun 130012, China
- Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun 130012, China
| | - Junqiu Liu
- State Key laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
- Correspondence: (J.L.); (S.G.); Tel.: +86-135-0432-8390 (S.G.)
| | - Shuwen Guan
- School of Life Sciences, Jilin University, Changchun 130012, China
- Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun 130012, China
- Correspondence: (J.L.); (S.G.); Tel.: +86-135-0432-8390 (S.G.)
| |
Collapse
|
26
|
Ranamalla SR, Porfire AS, Tomuță I, Banciu M. An Overview of the Supramolecular Systems for Gene and Drug Delivery in Tissue Regeneration. Pharmaceutics 2022; 14:pharmaceutics14081733. [PMID: 36015356 PMCID: PMC9412871 DOI: 10.3390/pharmaceutics14081733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/31/2022] [Accepted: 08/03/2022] [Indexed: 12/03/2022] Open
Abstract
Tissue regeneration is a prominent area of research, developing biomaterials aimed to be tunable, mechanistic scaffolds that mimic the physiological environment of the tissue. These biomaterials are projected to effectively possess similar chemical and biological properties, while at the same time are required to be safely and quickly degradable in the body once the desired restoration is achieved. Supramolecular systems composed of reversible, non-covalently connected, self-assembly units that respond to biological stimuli and signal cells have efficiently been developed as preferred biomaterials. Their biocompatibility and the ability to engineer the functionality have led to promising results in regenerative therapy. This review was intended to illuminate those who wish to envisage the niche translational research in regenerative therapy by summarizing the various explored types, chemistry, mechanisms, stimuli receptivity, and other advancements of supramolecular systems.
Collapse
Affiliation(s)
- Saketh Reddy Ranamalla
- Department of Pharmaceutical Technology and Bio Pharmacy, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400010 Cluj-Napoca, Romania
- Doctoral School in Integrative Biology, Faculty of Biology and Geology, “Babeș-Bolyai” University, 400015 Cluj-Napoca, Romania
| | - Alina Silvia Porfire
- Department of Pharmaceutical Technology and Bio Pharmacy, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400010 Cluj-Napoca, Romania
- Correspondence:
| | - Ioan Tomuță
- Department of Pharmaceutical Technology and Bio Pharmacy, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400010 Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, “Babeș-Bolyai” University, 400015 Cluj-Napoca, Romania
| |
Collapse
|
27
|
Wang X, Gao H, Zhang X, Qian S, Wang C, Deng L, Zhong M, Qing G. Aspartic Acid-Modified Phospholipids Regulate Cell Response and Rescue Memory Deficits in APP/PS1 Transgenic Mice. ACS Chem Neurosci 2022; 13:2154-2163. [PMID: 35818957 DOI: 10.1021/acschemneuro.2c00202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Misfolding and accumulation of amyloid-β (Aβ) to form senile plaques are the main neuropathological signatures of Alzheimer's disease (AD). Decreasing Aβ production, inhibiting Aβ aggregation, and clearing Aβ plaques are thus considered an important strategy for AD treatment. However, numerous drugs cannot enter the AD clinical trials due to unsatisfactory biocompatibility, poor blood-brain barrier penetration, little biomarker impact, and/or low therapeutic indicators. Here, a pair of chiral aspartic acid-modified 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine (l- and d-Asp-DPPE) are prepared to build stabilized chiral liposomes. We find that both l- and d-liposomes are able to rescue Aβ aggregation-induced apoptosis, oxidative stress, and calcium homeostasis, in which the effect of d-liposomes is more obvious than that of l-ones. Furthermore, in AD model mice (APPswe/PS1d9 double-transgenic mice), chiral liposomes not only show biosafety but also strongly improve cognitive deficits and reduce Aβ deposition in the brain. Our results suggest that chiral liposomes, particularly, d-liposomes, could be a potential therapeutic approach for AD treatment. This study opens new horizons by showing that liposomes will be used for drug development in addition to delivery and targeting functions.
Collapse
Affiliation(s)
- Xue Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P. R. China.,Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Huiling Gao
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, P. R. China
| | - Xiaoyu Zhang
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Shengxu Qian
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Cunli Wang
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| | - Lijing Deng
- Department of Critical Care Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu 610041, P. R. China
| | - Manli Zhong
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, P. R. China
| | - Guangyan Qing
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China
| |
Collapse
|
28
|
Preparation and pH/temperature dual drug release behavior of polyamino acid nanomicelles. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-021-03735-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
29
|
Liu XY, Zhang X, Yang JB, Wu CY, Wang Q, Lu ZL, Tang Q. Multifunctional amphiphilic peptide dendrimer as nonviral gene vectors for effective cancer therapy via combined gene/photodynamic therapies. Colloids Surf B Biointerfaces 2022; 217:112651. [PMID: 35759892 DOI: 10.1016/j.colsurfb.2022.112651] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 10/18/2022]
Abstract
Gene therapy holds great promise for treatment of gene-associated diseases. However, safe and successful clinical application urgently requires further advancement of constructing efficient delivery systems. Herein, three amphiphilic peptide dendrimers (TTC-L-KRR/KKK/KHH), containing the natural amino acid residues (lysine K, arginine R, and histidine H) and AIE-based photosensitizer (tetraphenylethenethiophene modified cyanoacrylate, TTC) modified with alkyl chain (L), have been designed and prepared for improving therapeutic potency via the combination of gene therapy (GT) and photodynamic therapy (PDT). All three compounds possessed typical aggregation-induced emission (AIE) characteristics and ultralow critical micelle concentrations (CMCs). The liposomes consisting of amphiphilic peptide dendrimers and dioleoylphosphatidylethanolamine (DOPE) can effectively bind DNA into nanoparticles with appropriate sizes, regular morphology and good biocompatibility. Among them, liposomes TTC-L-KKK/DOPE exhibited the highest transfection efficiency up to 5.7-fold as compared with Lipo2000 in HeLa cells. Meanwhile, rapid endocytosis, successful endo/lysosomal escape, gene release and rapid nuclear delivery of DNA revealed the superiority of liposomes TTC-L-KKK/DOPE during gene delivery process. More importantly, efficient reactive oxygen species (ROS) generation by TTC-L-KKK/DOPE led to effective PDT, thus improving therapeutic potency via combining with p53 mediated-gene therapy. Our work brought novel insight and direction for the construction of bio-safe and bio-imaging liposome as the multifunctional nonviral gene vectors for the effective combined gene/photodynamic therapies.
Collapse
Affiliation(s)
- Xu-Ying Liu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Xi Zhang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jing-Bo Yang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Cheng-Yan Wu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Qian Wang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Zhong-Lin Lu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China.
| | - Quan Tang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
30
|
Yan Y, Zhang G, Wu C, Ren Q, Liu X, Huang F, Cao Y, Ye W. Structural Exploration of Polycationic Nanoparticles for siRNA Delivery. ACS Biomater Sci Eng 2022; 8:1964-1974. [PMID: 35380797 DOI: 10.1021/acsbiomaterials.2c00196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
RNA interference (RNAi) is a promising approach to the treatment of genetic diseases by the specific knockdown of target genes. Functional polymers are potential vehicles for the effective delivery of vulnerable small interfering RNA (siRNA), which is required for the broad application of RNAi-based therapeutics. The development of methods for the facile modulation of chemical structures of polymeric carriers and an elucidation of detailed delivery mechanisms remain important areas of research. In this paper, we synthesized a series of methacrylate-based polymers with controllable structures and narrow distributions by atom transfer radical polymerization using various combinations of cationic monomers (2-dimethylaminoethyl methacrylate, 2-diethylaminoethyl methacrylate, and 2-dibutylaminoethyl methacrylate) and hydrophobic monomers (2-butyl methacrylate (BMA), cyclohexyl methacrylate, and 2-ethylhexyl methacrylate). These polymers exhibited varying hydrophobicities, charge densities, and pKa values, enabling the discovery of effective carriers for siRNA by in vitro delivery assays. For the polymers with BMA segments, 50% of cationic segments were beneficial to the formation of siRNA nanoparticles (NPs) and the in vitro delivery of siRNA. The optimal ratio varied for different combinations of cationic and hydrophobic segments. In particular, 20k PMB 0.5, PME 0.5, and PEB 1.0 showed >75% luciferase knockdown. Efficacious delivery was dependent on high siRNA binding, the small size of NPs, and balanced hydrophobicity and charge density. Cellular uptake and endosomal escape experiments indicated that carboxybetaine modification of 20k PMB 0.5 did not remarkably affect the internalization of corresponding NPs after incubation for 6 h but significantly reduced the endosomal escape of NPs, which leads to the notable decrease in delivery efficacy of polymers. These results provide insights into the mechanism of polymer-based siRNA delivery and may inspire the development of novel polymeric carriers.
Collapse
Affiliation(s)
- Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Guangliang Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Chengfan Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Qidi Ren
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Xiaomin Liu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Fangqian Huang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yi Cao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Wenbo Ye
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
31
|
Rodríguez‐Izquierdo I, Sepúlveda‐Crespo D, Lasso JM, Resino S, Muñoz‐Fernández MÁ. Baseline and time-updated factors in preclinical development of anionic dendrimers as successful anti-HIV-1 vaginal microbicides. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1774. [PMID: 35018739 PMCID: PMC9285063 DOI: 10.1002/wnan.1774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/05/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022]
Abstract
Although a wide variety of topical microbicides provide promising in vitro and in vivo efficacy, most of them failed to prevent sexual transmission of human immunodeficiency virus type 1 (HIV-1) in human clinical trials. In vitro, ex vivo, and in vivo models must be optimized, considering the knowledge acquired from unsuccessful and successful clinical trials to improve the current gaps and the preclinical development protocols. To date, dendrimers are the only nanotool that has advanced to human clinical trials as topical microbicides to prevent HIV-1 transmission. This fact demonstrates the importance and the potential of these molecules as microbicides. Polyanionic dendrimers are highly branched nanocompounds with potent activity against HIV-1 that disturb HIV-1 entry. Herein, the most significant advancements in topical microbicide development, trying to mimic the real-life conditions as closely as possible, are discussed. This review also provides the preclinical assays that anionic dendrimers have passed as microbicides because they can improve current antiviral treatments' efficacy. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
| | - Daniel Sepúlveda‐Crespo
- Unidad de Infección Viral e Inmunidad, Centro Nacional de MicrobiologíaInstituto de Salud Carlos IIIMadridSpain
| | | | - Salvador Resino
- Unidad de Infección Viral e Inmunidad, Centro Nacional de MicrobiologíaInstituto de Salud Carlos IIIMadridSpain
| | - Ma Ángeles Muñoz‐Fernández
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM)MadridSpain
- Spanish HIV HGM BioBankMadridSpain
- Section of Immunology, Laboratorio InmunoBiología MolecularHospital General Universitario Gregorio Marañón (HGUGM)MadridSpain
| |
Collapse
|
32
|
Wang J, Chen G, Liu N, Han X, Zhao F, Zhang L, Chen P. Strategies for improving the safety and RNAi efficacy of noncovalent peptide/siRNA nanocomplexes. Adv Colloid Interface Sci 2022; 302:102638. [PMID: 35299136 DOI: 10.1016/j.cis.2022.102638] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 12/12/2022]
Abstract
In the past decades, the striking development of cationic polypeptides and cell-penetrating peptides (CPPs) tailored for small interfering RNA (siRNA) delivery has been fuelled by the conception of nuclear acid therapy and precision medicine. Owing to their amino acid compositions, inherent secondary structures as well as diverse geometrical shapes, peptides or peptide-containing polymers exhibit good biodegradability, high flexibility, and bio-functional diversity as nonviral siRNA vectors. Also, a variety of noncovalent nanocomplexes could be built via self-assembling and electrostatic interactions between cationic peptides and siRNAs. Although the peptide/siRNA nanocomplex-based RNAi therapies, STP705 and MIR-19, are under clinical trials, a guideline addressing the current bottlenecks of peptide/siRNA nanocomplex delivery is in high demand for future research and development. In this review, we present strategies for improving the safety and RNAi efficacy of noncovalent peptide/siRNA nanocomplexes in the treatment of genetic disorders. Through thorough analysis of those RNAi formulations using different delivery strategies, we seek to shed light on the rationale of peptide design and modification in constructing robust siRNA delivery systems, including targeted and co-delivery systems. Based on this, we provide a timely and comprehensive understanding of how to engineer biocompatible and efficient peptide-based siRNA vectors.
Collapse
Affiliation(s)
- Jun Wang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Guang Chen
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada; Key Laboratory of Chemical Additives for China National Light Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Nan Liu
- Advanced Materials Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250014, China
| | - Xiaoxia Han
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Feng Zhao
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - P Chen
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada; Advanced Materials Institute, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250014, China.
| |
Collapse
|
33
|
Halib N, Pavan N, Trombetta C, Dapas B, Farra R, Scaggiante B, Grassi M, Grassi G. An Overview of siRNA Delivery Strategies for Urological Cancers. Pharmaceutics 2022; 14:718. [PMID: 35456552 PMCID: PMC9030829 DOI: 10.3390/pharmaceutics14040718] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/18/2022] [Accepted: 03/24/2022] [Indexed: 02/05/2023] Open
Abstract
The treatment of urological cancers has been significantly improved in recent years. However, for the advanced stages of these cancers and/or for those developing resistance, novel therapeutic options need to be developed. Among the innovative strategies, the use of small interfering RNA (siRNA) seems to be of great therapeutic interest. siRNAs are double-stranded RNA molecules which can specifically target virtually any mRNA of pathological genes. For this reason, siRNAs have a great therapeutic potential for human diseases including urological cancers. However, the fragile nature of siRNAs in the biological environment imposes the development of appropriate delivery systems to protect them. Thus, ensuring siRNA reaches its deep tissue target while maintaining structural and functional integrity represents one of the major challenges. To reach this goal, siRNA-based therapies require the development of fine, tailor-made delivery systems. Polymeric nanoparticles, lipid nanoparticles, nanobubbles and magnetic nanoparticles are among nano-delivery systems studied recently to meet this demand. In this review, after an introduction about the main features of urological tumors, we describe siRNA characteristics together with representative delivery systems developed for urology applications; the examples reported are subdivided on the basis of the different delivery materials and on the different urological cancers.
Collapse
Affiliation(s)
- Nadia Halib
- Department of Basic Sciences & Oral Biology, Faculty of Dentistry, Universiti Sains Islam Malaysia, Kuala Lumpur 55100, Malaysia;
| | - Nicola Pavan
- Urology Clinic, Department of Medical, Surgical and Health Science, University of Trieste, I-34149 Trieste, Italy; (N.P.); (C.T.)
| | - Carlo Trombetta
- Urology Clinic, Department of Medical, Surgical and Health Science, University of Trieste, I-34149 Trieste, Italy; (N.P.); (C.T.)
| | - Barbara Dapas
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy; (B.D.); (R.F.); (B.S.)
| | - Rossella Farra
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy; (B.D.); (R.F.); (B.S.)
| | - Bruna Scaggiante
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy; (B.D.); (R.F.); (B.S.)
| | - Mario Grassi
- Department of Engineering and Architecture, Trieste University, Via Valerio 6, I-34127 Trieste, Italy;
| | - Gabriele Grassi
- Department of Life Sciences, Cattinara University Hospital, Trieste University, Strada di Fiume 447, I-34149 Trieste, Italy; (B.D.); (R.F.); (B.S.)
| |
Collapse
|
34
|
Byun MJ, Lim J, Kim SN, Park DH, Kim TH, Park W, Park CG. Advances in Nanoparticles for Effective Delivery of RNA Therapeutics. BIOCHIP JOURNAL 2022; 16:128-145. [PMID: 35261724 PMCID: PMC8891745 DOI: 10.1007/s13206-022-00052-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/24/2022] [Accepted: 02/06/2022] [Indexed: 12/17/2022]
Abstract
RNA therapeutics, including messenger RNA (mRNA) and small interfering RNA (siRNA), are genetic materials that mediate the translation of genetic direction from genes to induce or inhibit specific protein production. Although the interest in RNA therapeutics is rising globally, the absence of an effective delivery system is an obstacle to the clinical application of RNA therapeutics. Additionally, immunogenicity, short duration of protein expression, unwanted enzymatic degradation, and insufficient cellular uptake could limit the therapeutic efficacy of RNA therapeutics. In this regard, novel platforms based on nanoparticles are crucial for delivering RNAs to the targeted site to increase efficiency without toxicity. In this review, the most recent status of nanoparticles as RNA delivery vectors, with a focus on polymeric nanoparticles, peptide-derived nanoparticles, inorganic nanoparticles, and hybrid nanoparticles, is discussed. These nanoparticular platforms can be utilized for safe and effective RNA delivery to augment therapeutic effects. Ultimately, RNA therapeutics encapsulated in nanoparticle-based carriers will be used to treat many diseases and save lives.
Collapse
Affiliation(s)
- Min Ji Byun
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
| | - Jaesung Lim
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
| | - Se-Na Kim
- Institute of Medical and Biological Engineering, Medical Research Center, Seoul National University, Seoul, 03080 Republic of Korea
| | - Dae-Hwan Park
- Department of Engineering Chemistry, Chungbuk National University, Cheongju, Chungbuk 28644 Republic of Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, 84, Heukseok-ro, Dongjak-gu, Seoul, 06974 Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419 Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Gyeonggi 16419 Republic of Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Suwon, Gyeonggi 16419 Republic of Korea
| |
Collapse
|
35
|
Ashrafizadeh M, Zarrabi A, Mostafavi E, Aref AR, Sethi G, Wang L, Tergaonkar V. Non-coding RNA-based regulation of inflammation. Semin Immunol 2022; 59:101606. [PMID: 35691882 DOI: 10.1016/j.smim.2022.101606] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 05/01/2022] [Accepted: 05/25/2022] [Indexed: 01/15/2023]
Abstract
Inflammation is a multifactorial process and various biological mechanisms and pathways participate in its development. The presence of inflammation is involved in pathogenesis of different diseases such as diabetes mellitus, cardiovascular diseases and even, cancer. Non-coding RNAs (ncRNAs) comprise large part of transcribed genome and their critical function in physiological and pathological conditions has been confirmed. The present review focuses on miRNAs, lncRNAs and circRNAs as ncRNAs and their potential functions in inflammation regulation and resolution. Pro-inflammatory and anti-inflammatory factors are regulated by miRNAs via binding to 3'-UTR or indirectly via affecting other pathways such as SIRT1 and NF-κB. LncRNAs display a similar function and they can also affect miRNAs via sponging in regulating levels of cytokines. CircRNAs mainly affect miRNAs and reduce their expression in regulating cytokine levels. Notably, exosomal ncRNAs have shown capacity in inflammation resolution. In addition to pre-clinical studies, clinical trials have examined role of ncRNAs in inflammation-mediated disease pathogenesis and cytokine regulation. The therapeutic targeting of ncRNAs using drugs and nucleic acids have been analyzed to reduce inflammation in disease therapy. Therefore, ncRNAs can serve as diagnostic, prognostic and therapeutic targets in inflammation-related diseases in pre-clinical and clinical backgrounds.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396 Istanbul, Turkey.
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Translational Sciences, Xsphera Biosciences Inc. 6, Tide Street, Boston, MA 02210, USA
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore.
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signaling, Institute of Molecular and Cell Biology (IMCB), Singapore, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
36
|
Amjad H, Rizwan K, Baber M, Iqbal S. Organic–Inorganic Nanohybrids in Cancer Treatment. MATERIALS HORIZONS: FROM NATURE TO NANOMATERIALS 2022:107-132. [DOI: 10.1007/978-981-19-4538-0_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
37
|
Puzari M, Chetia P. Nanotechnology-based cancer drug delivery. ADVANCES IN NANOTECHNOLOGY-BASED DRUG DELIVERY SYSTEMS 2022:415-422. [DOI: 10.1016/b978-0-323-88450-1.00007-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
38
|
Chauhan S, Jain K, Naqvi S. Dendrimers and its theranostic applications in infectious diseases. NANOTHERANOSTICS FOR TREATMENT AND DIAGNOSIS OF INFECTIOUS DISEASES 2022:199-228. [DOI: 10.1016/b978-0-323-91201-3.00004-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
39
|
Apartsin E, Caminade A. Supramolecular Self-Associations of Amphiphilic Dendrons and Their Properties. Chemistry 2021; 27:17976-17998. [PMID: 34713506 PMCID: PMC9298340 DOI: 10.1002/chem.202102589] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Indexed: 12/15/2022]
Abstract
This review presents precisely defined amphiphilic dendrons, their self-association properties, and their different uses. Dendrons, also named dendritic wedges, are composed of a core having two different types of functions, of which one type is used for growing or grafting branched arms, generally multiplied by 2 at each layer by using 1→2 branching motifs. A large diversity of structures has been already synthesized. In practically all cases, their synthesis is based on the synthesis of known dendrimers, such as poly(aryl ether), poly(amidoamine) (in particular PAMAM), poly(amide) (in particular poly(L-lysine)), 1→3 branching motifs (instead of 1→2), poly(alkyl ether) (poly(glycerol) and poly(ethylene glycol)), poly(ester), and those containing main group elements (poly(carbosilane) and poly(phosphorhydrazone)). In most cases, the hydrophilic functions are on the surface of the dendrons, whereas one or two hydrophobic tails are linked to the core. Depending on the structure of the dendrons, and on the experimental conditions used, the amphiphilic dendrons can self-associate at the air-water interface, or form micelles (eventually tubular, but most generally spherical), or form vesicles. These associated dendrons are suitable for the encapsulation of low-molecular or macromolecular bioactive entities to be delivered in cells. This review is organized depending on the nature of the internal structure of the amphiphilic dendrons (aryl ether, amidoamine, amide, quaternary carbon atom, alkyl ether, ester, main group element). The properties issued from their self-associations are described all along the review.
Collapse
Affiliation(s)
- Evgeny Apartsin
- Laboratoire de Chimie de Coordination (LCC) CNRS205 route de Narbonne31077Toulouse cedex 4France
- LCC-CNRSUniversité de Toulouse, CNRS31077Toulouse cedex 4France
- Institute of Chemical Biology and Fundamental Medicine630090NovosibirskRussia
- Novosibirsk State University630090NovosibirskRussia
| | - Anne‐Marie Caminade
- Laboratoire de Chimie de Coordination (LCC) CNRS205 route de Narbonne31077Toulouse cedex 4France
- LCC-CNRSUniversité de Toulouse, CNRS31077Toulouse cedex 4France
| |
Collapse
|
40
|
Wang R, Yin C, Liu C, Sun Y, Xiao P, Li J, Yang S, Wu W, Jiang X. Phenylboronic Acid Modification Augments the Lysosome Escape and Antitumor Efficacy of a Cylindrical Polymer Brush-Based Prodrug. J Am Chem Soc 2021; 143:20927-20938. [PMID: 34855390 DOI: 10.1021/jacs.1c09741] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Timely lysosome escape is of paramount importance for endocytosed nanomedicines to avoid premature degradation under the acidic and hydrolytic conditions in lysosomes. Herein, we report an exciting finding that phenylboronic acid (PBA) modification can greatly facilitate the lysosome escape of cylindrical polymer brushes (CPBs). On the basis of our experimental results, we speculate that the mechanism is associated with the specific interactions of the PBA groups with lysosomal membrane proteins and hot shock proteins. The featured advantage of the PBA modification over the known lysosome escape strategies is that it does not cause significant adverse effects on the properties of the CPBs; on the contrary, it enhances remarkably their tumor accumulation and penetration. Furthermore, doxorubicin was conjugated to the PBA-modified CPBs with a drug loading content larger than 20%. This CPBs-based prodrug could eradicate the tumors established in mice by multiple intravenous administrations. This work provides a novel strategy for facilitating the lysosome escape of nanomaterials and demonstrates that PBA modification is an effective way to improve the overall properties of nanomedicines including the tumor therapeutic efficacy.
Collapse
Affiliation(s)
- Ruonan Wang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Changfeng Yin
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Changren Liu
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Ying Sun
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Panpan Xiao
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Jia Li
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Shuo Yang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Wei Wu
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Xiqun Jiang
- Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
41
|
Amani A, Dustparast M, Noruzpour M, Zakaria RA, Ebrahimi HA. Design and Invitro Characterization of Green Synthesized Magnetic Nanoparticles Conjugated with Multitargeted Poly Lactic Acid Copolymers for Co-delivery of siRNA and Paclitaxel. Eur J Pharm Sci 2021; 167:106007. [PMID: 34520835 DOI: 10.1016/j.ejps.2021.106007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/21/2021] [Accepted: 09/10/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND The self-assembling of various amphipathic copolymers is a simple method that allows the preparation of complex nanoparticles with several useful properties. In the present study, the polylactic acid-polyethylene glycol-folate (PLA-PEG-FA) (PPF), PLA-PEG-T7 peptide (PPT) and PLA-Chitosan-Spermine (PCS) copolymers were synthesized separately. METHODS These copolymers combined with Fe3O4 magnetic core and loaded with paclitaxel (PTX)/siRNA-FAM to form magnetic PCS/PPF/PPT/PTX/siRNA micelles (MPCSFT/PTX/siRNA) and were characterized using physicochemical and biological analysis. RESULTS The results revealed that the MPCSPFT/PTX/siRNA had spherical morphology with particle size and zeta potential about 197 nm and -7.8 mV, respectively. Release assay was determined under neutral (pH=7.4) and acidic pH (pH=6) conditions to simulate PTX and siRNA release profile from MPCSPFT/PTX/siRNA micelles in normal and cancerous tissues. The ability of MPCSPFT for co-delivery of PTX and siRNA into MCF-7 cells was determined by MTT and flow cytometry tests, respectively. The results revealed that the release rate of siRNA and PTX from MPCSPFT/PTX/siRNA nanoparticles under an acidic environment (pH=6) was significantly higher than that of their release rate in a neutral medium (pH=7.4). CONCLUSION Conjugation of both folic acid and T7-peptide on the surface of micelles compared to separate conjugation of one of these ligands, increased the efficiency of drug and siRNA delivery to breast cancer cells.
Collapse
Affiliation(s)
- Amin Amani
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Mohammad Dustparast
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mehran Noruzpour
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Rasool Asghari Zakaria
- Department of Agronomy and Plant Breeding, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Hossein Ali Ebrahimi
- Department of Pharmaceutics, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
42
|
Pre-Clinical and Clinical Applications of Small Interfering RNAs (siRNA) and Co-Delivery Systems for Pancreatic Cancer Therapy. Cells 2021; 10:cells10123348. [PMID: 34943856 PMCID: PMC8699513 DOI: 10.3390/cells10123348] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer (PC) is one of the leading causes of death and is the fourth most malignant tumor in men. The epigenetic and genetic alterations appear to be responsible for development of PC. Small interfering RNA (siRNA) is a powerful genetic tool that can bind to its target and reduce expression level of a specific gene. The various critical genes involved in PC progression can be effectively targeted using diverse siRNAs. Moreover, siRNAs can enhance efficacy of chemotherapy and radiotherapy in inhibiting PC progression. However, siRNAs suffer from different off target effects and their degradation by enzymes in serum can diminish their potential in gene silencing. Loading siRNAs on nanoparticles can effectively protect them against degradation and can inhibit off target actions by facilitating targeted delivery. This can lead to enhanced efficacy of siRNAs in PC therapy. Moreover, different kinds of nanoparticles such as polymeric nanoparticles, lipid nanoparticles and metal nanostructures have been applied for optimal delivery of siRNAs that are discussed in this article. This review also reveals that how naked siRNAs and their delivery systems can be exploited in treatment of PC and as siRNAs are currently being applied in clinical trials, significant progress can be made by translating the current findings into the clinical settings.
Collapse
|
43
|
Pan X, Veroniaina H, Su N, Sha K, Jiang F, Wu Z, Qi X. Applications and developments of gene therapy drug delivery systems for genetic diseases. Asian J Pharm Sci 2021; 16:687-703. [PMID: 35027949 PMCID: PMC8737406 DOI: 10.1016/j.ajps.2021.05.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 02/15/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Genetic diseases seriously threaten human health and have always been one of the refractory conditions facing humanity. Currently, gene therapy drugs such as siRNA, shRNA, antisense oligonucleotide, CRISPR/Cas9 system, plasmid DNA and miRNA have shown great potential in biomedical applications. To avoid the degradation of gene therapy drugs in the body and effectively deliver them to target tissues, cells and organelles, the development of excellent drug delivery vehicles is of utmost importance. Viral vectors are the most widely used delivery vehicles for gene therapy in vivo and in vitro due to their high transfection efficiency and stable transgene expression. With the development of nanotechnology, novel nanocarriers are gradually replacing viral vectors, emerging superior performance. This review mainly illuminates the current widely used gene therapy drugs, summarizes the viral vectors and non-viral vectors that deliver gene therapy drugs, and sums up the application of gene therapy to treat genetic diseases. Additionally, the challenges and opportunities of the field are discussed from the perspective of developing an effective nano-delivery system.
Collapse
Affiliation(s)
- Xiuhua Pan
- China Pharmaceutical University, Nanjing 211198, China
| | | | - Nan Su
- China Pharmaceutical University, Nanjing 211198, China
| | - Kang Sha
- China Pharmaceutical University, Nanjing 211198, China
| | - Fenglin Jiang
- China Pharmaceutical University, Nanjing 211198, China
| | - Zhenghong Wu
- China Pharmaceutical University, Nanjing 211198, China
| | - Xiaole Qi
- China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
44
|
Zhao C, Chen H, Wang F, Zhang X. Amphiphilic self-assembly peptides: Rational strategies to design and delivery for drugs in biomedical applications. Colloids Surf B Biointerfaces 2021; 208:112040. [PMID: 34425532 DOI: 10.1016/j.colsurfb.2021.112040] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/21/2021] [Accepted: 08/12/2021] [Indexed: 01/01/2023]
Abstract
Amphiphilic self-assembling peptides are widely used in tissue and cell engineering, antimicrobials, drug-delivery systems and other biomedical fields due to their good biocompatibility, functionality, flexibility of design and synthesis, and tremendous potential as delivery carriers for drugs. Currently, the design and study of amphipathic peptides by a bottom-up method to develop new biomedical materials have become a hot topic. However, defined rules have not been established for the design and development of self-assembled peptides. Therefore, the focus of this review is to summarize and provide several rational strategies for the design and study of amphiphilic self-assembly peptides. In addition, this paper also describes the types and general self-assembling mechanism of amphipathic peptides, and outlines their applications in the delivery of hydrophobic drugs, nucleic acid drugs, peptide drugs and vaccines. Amphiphilic self-assembled peptides are expected to exploit new functional materials for drug delivery and other applications.
Collapse
Affiliation(s)
- Chunqian Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, People's Republic of China.
| | - Hongyuan Chen
- Department of General Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong University, Jinan, 250021, People's Republic of China.
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, People's Republic of China.
| | - Xinke Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, People's Republic of China.
| |
Collapse
|
45
|
A Self-Assembling Amphiphilic Peptide Dendrimer-Based Drug Delivery System for Cancer Therapy. Pharmaceutics 2021; 13:pharmaceutics13071092. [PMID: 34371783 PMCID: PMC8309127 DOI: 10.3390/pharmaceutics13071092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/08/2021] [Accepted: 07/14/2021] [Indexed: 12/24/2022] Open
Abstract
Despite being a mainstay of clinical cancer treatment, chemotherapy is limited by its severe side effects and inherent or acquired drug resistance. Nanotechnology-based drug-delivery systems are widely expected to bring new hope for cancer therapy. These systems exploit the ability of nanomaterials to accumulate and deliver anticancer drugs at the tumor site via the enhanced permeability and retention effect. Here, we established a novel drug-delivery nanosystem based on amphiphilic peptide dendrimers (AmPDs) composed of a hydrophobic alkyl chain and a hydrophilic polylysine dendron with different generations (AmPD KK2 and AmPD KK2K4). These AmPDs assembled into nanoassemblies for efficient encapsulation of the anti-cancer drug doxorubicin (DOX). The AmPDs/DOX nanoformulations improved the intracellular uptake and accumulation of DOX in drug-resistant breast cancer cells and increased permeation in 3D multicellular tumor spheroids in comparison with free DOX. Thus, they exerted effective anticancer activity while circumventing drug resistance in 2D and 3D breast cancer models. Interestingly, AmPD KK2 bearing a smaller peptide dendron encapsulated DOX to form more stable nanoparticles than AmPD KK2K4 bearing a larger peptide dendron, resulting in better cellular uptake, penetration, and anti-proliferative activity. This may be because AmPD KK2 maintains a better balance between hydrophobicity and hydrophilicity to achieve optimal self-assembly, thereby facilitating more stable drug encapsulation and efficient drug release. Together, our study provides a promising perspective on the design of the safe and efficient cancer drug-delivery nanosystems based on the self-assembling amphiphilic peptide dendrimer.
Collapse
|
46
|
Lithocholic acid-tryptophan conjugate (UniPR126) based mixed micelle as a nano carrier for specific delivery of niclosamide to prostate cancer via EphA2 receptor. Int J Pharm 2021; 605:120819. [PMID: 34166727 DOI: 10.1016/j.ijpharm.2021.120819] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 12/25/2022]
Abstract
Targeted delivery of chemotherapeutic agents is considered a prominent strategy for the treatment of cancer due to its site-specific delivery, augmented penetration, bioavailability, and improved therapeutic efficiency. In the present study, we employed UniPR126 as a carrier in a mixed nanomicellar delivery system to target and deliver anticancer drug NIC specifically to cancer cells via EphA2 receptors as these receptors are overexpressed in cancer cells but not in normal cells. The specificity of the carrier was confirmed from the significant enhancement in the uptake of coumarin-6 loaded mixed nanomicelle by EphA2 highly expressed PC-3 cells compared to EphA2 low expressed H4 cells. Further, niclosamide-loaded lithocholic acid tryptophan conjugate-based mixed nanomicelle has shown significant synergistic cytotoxicity in PC-3 but not in H4 cells. In vivo anticancer efficacy data in PC-3 xenograft revealed a significant reduction in the tumor volume (66.87%) with niclosamide-loaded lithocholic acid tryptophan conjugate nanomicelle, where pure niclosamide showed just half of the activity. Molecular signaling data by western blotting also indicated that niclosamide-loaded lithocholic acid tryptophan conjugate nanomicelle interfered with the EphA2 receptor signaling and inhibition of the Wnt/beta-catenin pathway and resulted in the synergistic anticancer activity compared to niclosamide pure drug.
Collapse
|
47
|
Wu Y, Zhong D, Li Y, Wu H, Zhang H, Mao H, Yang J, Luo K, Gong Q, Gu Z. A tumor-activatable peptide supramolecular nanoplatform for the delivery of dual-gene targeted siRNAs for drug-resistant cancer treatment. NANOSCALE 2021; 13:4887-4898. [PMID: 33625408 DOI: 10.1039/d0nr08487e] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Combinatorial short interference RNA (siRNA) technology for the silencing of multiple genes is expected to provide an effective therapeutic approach for cancer with complex genetic mutation and dysregulation. Herein we present a tumor-activatable supramolecular nanoplatform for the delivery of siRNAs to target telomerase and telomeres for paclitaxel-resistant non-small-cell lung cancer (A549/PTX) treatment. Two different sequences of siRNA are incorporated in a single nanoparticle, which is obtained by self-assembly from a peptide dendrimer. The siRNA stability is improved by the nanoparticle in the presence of serum compared to free siRNA, and these siRNAs are protected from RNA enzyme degradation. In the tumor extracellular acid environment, the PEG corona of the nanoparticle is removed to promote the internalization of siRNAs into tumor cells. The disulfide linkages between the nanoparticle and siRNAs are cleared in the reductive environment of the tumor cells, and the siRNAs are released in the cytoplasm. In vitro experiments show that the gene expression of hTERT and TRF2 at the mRNA and protein levels of A549/PTX tumor cells is down-regulated, which results in cooperative restraining proliferation and invasion of A549/PTX tumor cells. For the tumor cell-targeting function of the MUC1 aptamer and the EPR effect, sufficient tumor accumulation of nanoparticles was observed. Meanwhile, a shift of negative surface charge of nanoparticles to positive charge in the tumor extracellular microenvironment enhances deep penetration of siRNA-incorporating nanoparticles into tumor tissues. In vivo animal studies support that successful down-regulation of hTERT and TRF2 gene expression achieves effective inhibition of the growth and neovascularization of drug-resistant tumor cells. This work has provided a new avenue for drug-resistant cancer treatment by designing and synthesizing a tumor-activatable nanoplatform to achieve the delivery of dual-gene targeted combinatorial siRNAs.
Collapse
Affiliation(s)
- Yahui Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, P. R. China.
| | - Dan Zhong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, P. R. China.
| | - Yunkun Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, P. R. China.
| | - Huayu Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, P. R. China.
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, CA 91711, USA
| | - Hongli Mao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816, P. R. China
| | - Jun Yang
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P. R. China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, P. R. China.
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, P. R. China.
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610041, P. R. China. and Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, NJTech-BARTY Joint Research Center for Innovative Medical Technology, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816, P. R. China
| |
Collapse
|
48
|
Saviano F, Lovato T, Russo A, Russo G, Bouton CR, Shattock RJ, Alexander C, Quaglia F, Blakney AK, Gurnani P, Conte C. Ornithine-derived oligomers and dendrimers for in vitro delivery of DNA and ex vivo transfection of skin cells via saRNA. J Mater Chem B 2021; 8:4940-4949. [PMID: 32463058 DOI: 10.1039/d0tb00942c] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gene therapies are undergoing a renaissance, primarily due to their potential for applications in vaccination for infectious diseases and cancers. Although the biology of these technologies is rapidly evolving, delivery strategies need to be improved to overcome the poor pharmacokinetics and cellular transport of nucleic acids whilst maintaining patient safety. In this work, we describe the divergent synthesis of biodegradable cationic dendrimers based on the amino acid ornithine as non-viral gene delivery vectors and evaluate their potential as delivery vectors for DNA and RNA. The dendrimers effectively complexed model nucleic acids at lower N/P ratios than polyethyleneimine and outperformed it in DNA transfection experiments with ratios above 5. Remarkably, all dendrimer polyplexes at N/P = 2 achieved up to 7-fold higher protein content over an optimized PEI formulation when used for transfections with self-amplifying RNA (saRNA). Finally, transfection studies utilizing human skin explants revealed an increase of cells producing protein from 2% with RNA alone to 12% with dendrimer polyplexes, attributed to expression enrichment predominantly in epithelial cells, fibroblasts and leukocytes, with minor enrichment in NK cells, T cells, monocytes, and B cells. Overall, this study indicates the clear potential of ornithine dendrimers as safe and effective delivery vectors for both DNA and RNA therapeutics.
Collapse
Affiliation(s)
- Francesca Saviano
- Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy.
| | - Tatiana Lovato
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK.
| | - Annapina Russo
- Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy.
| | - Giulia Russo
- Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy.
| | - Clément R Bouton
- Department of Infectious Disease, Imperial College London, School of Medicine, St Mary's Hospital, Praed Street, London W2 1NY, UK.
| | - Robin J Shattock
- Department of Infectious Disease, Imperial College London, School of Medicine, St Mary's Hospital, Praed Street, London W2 1NY, UK.
| | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK.
| | - Fabiana Quaglia
- Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy.
| | - Anna K Blakney
- Department of Infectious Disease, Imperial College London, School of Medicine, St Mary's Hospital, Praed Street, London W2 1NY, UK.
| | - Pratik Gurnani
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, NG7 2RD, UK.
| | - Claudia Conte
- Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy.
| |
Collapse
|
49
|
Folliero V, Zannella C, Chianese A, Stelitano D, Ambrosino A, De Filippis A, Galdiero M, Franci G, Galdiero M. Application of Dendrimers for Treating Parasitic Diseases. Pharmaceutics 2021; 13:343. [PMID: 33808016 PMCID: PMC7998910 DOI: 10.3390/pharmaceutics13030343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 01/02/2023] Open
Abstract
Despite advances in medical knowledge, parasitic diseases remain a significant global health burden and their pharmacological treatment is often hampered by drug toxicity. Therefore, drug delivery systems may provide useful advantages when used in combination with conventional therapeutic compounds. Dendrimers are three-dimensional polymeric structures, characterized by a central core, branches and terminal functional groups. These nanostructures are known for their defined structure, great water solubility, biocompatibility and high encapsulation ability against a wide range of molecules. Furthermore, the high ratio between terminal groups and molecular volume render them a hopeful vector for drug delivery. These nanostructures offer several advantages compared to conventional drugs for the treatment of parasitic infection. Dendrimers deliver drugs to target sites with reduced dosage, solving side effects that occur with accepted marketed drugs. In recent years, extensive progress has been made towards the use of dendrimers for therapeutic, prophylactic and diagnostic purposes for the management of parasitic infections. The present review highlights the potential of several dendrimers in the management of parasitic diseases.
Collapse
Affiliation(s)
- Veronica Folliero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (C.Z.); (A.C.); (D.S.); (A.A.); (M.G.)
| | - Carla Zannella
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (C.Z.); (A.C.); (D.S.); (A.A.); (M.G.)
| | - Annalisa Chianese
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (C.Z.); (A.C.); (D.S.); (A.A.); (M.G.)
| | - Debora Stelitano
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (C.Z.); (A.C.); (D.S.); (A.A.); (M.G.)
| | - Annalisa Ambrosino
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (C.Z.); (A.C.); (D.S.); (A.A.); (M.G.)
| | - Anna De Filippis
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Marilena Galdiero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (C.Z.); (A.C.); (D.S.); (A.A.); (M.G.)
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy;
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.F.); (C.Z.); (A.C.); (D.S.); (A.A.); (M.G.)
| |
Collapse
|
50
|
Hall R, Alasmari A, Mozaffari S, Mahdipoor P, Parang K, Montazeri Aliabadi H. Peptide/Lipid-Associated Nucleic Acids (PLANAs) as a Multicomponent siRNA Delivery System. Mol Pharm 2021; 18:986-1002. [PMID: 33496597 DOI: 10.1021/acs.molpharmaceut.0c00969] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RNAi is a biological process that utilizes small interfering RNA (siRNA) to prevent the translation of mRNA to protein. This mechanism could be beneficial in preventing the overexpression of proteins in cancer. However, the cellular delivery of siRNA has proven to be challenging due to its inherent negative charge and relative instability. Here, we designed a multicomponent delivery system composed of a specifically designed peptide (linear or cyclic fatty acyl peptide conjugates and hybrid cyclic/linear peptides) and several lipids (DOTAP, DOPE, cholesterol, and phosphatidylcholine) to form a nanoparticle, which we have termed as peptide lipid-associated nucleic acids (PLANAs). Five formulations were prepared (a formulation with no peptide, which was named lipid-associated nucleic acid or LANA, and PLANA formulations A-D) using a mini extruder to form uniform nanoparticles around 100 nm in size with a slightly positive charge (less than +10 mv). Formulations were evaluated for peptide incorporation, siRNA encapsulation efficiency, release profile, toxicity, cellular uptake, and protein silencing. Our experiments showed effective encapsulation of siRNA (>95%), a controlled release profile, and negligible toxicity in formulations that did not contain a positively charged lipid. The results also revealed that PLANAs C and D exhibited optimum cellular uptake (with 80-90% siRNA-positive cells for most of the formulations). PLANA D formulation was selected to silence two model proteins (Src and RPS6KA5) in the triple-negative human breast cancer cell line MDA-MB-231, with promising silencing efficiency, which diminished the expression of RPS6KA5 and Src to approximately 29 and 38% compared to naïve cells, respectively. Many approaches have been investigated for safe and efficient delivery of nucleic acids in the last 20 years; however, many have failed due to the multifaceted challenges to overcome. Our results show a promising potential for a multicomponent design that incorporates different components for a variety of delivery tasks, which warrants further investigation of PLANAs in vivo.
Collapse
Affiliation(s)
- Ryley Hall
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Abdulaziz Alasmari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Saghar Mozaffari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Parvin Mahdipoor
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| | - Hamidreza Montazeri Aliabadi
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, California 92618, United States
| |
Collapse
|