1
|
Wu Y, Kawamoto Y, Sun J, Takahashi Y, Higuchi Y, Takakura Y. Improvement of Drug Release from an Aptamer Drug Conjugate Using Reductive-sensitive Linkers for Tumor-targeted Drug Delivery. AAPS J 2025; 27:95. [PMID: 40397061 DOI: 10.1208/s12248-025-01070-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/04/2025] [Indexed: 05/22/2025] Open
Abstract
The selective delivery of small molecule compounds such as Gemcitabine to tumor cells is a promising methodology for enhancing therapeutic efficacy and attenuating the side effects of anticancer drugs. Aptamers are useful as target-directed ligands for tumor-selective drug delivery due to their ability to bind specific proteins. However, the drug must be released from the aptamer after the conjugate is taken up by the cell to exert its pharmacological effect. In this study, we designed and synthesized a conjugate in which a linker cleaved by glutathione, which is highly expressed in tumor cells, was inserted between the aptamer (AS1411) and Gemcitabine. Almost all Gemcitabine was released from the conjugate after 30 min in the presence of 6 mM glutathione. AS1411 is known to bind to nucleolin, which is highly expressed on tumor cells. The cytotoxicity of the AS1411 and Gemcitabine conjugate with a disulfide bond on A549 cells was higher than that of the conjugate without a disulfide bond. Furthermore, the cytotoxicity of the disulfide-linked conjugate of AS1411 and Gemcitabine was higher in A549 cells than in MCF10A cells, which were used as the model of normal cells. These results indicate that disulfide conjugation enhanced the tumor cell-selective cytotoxicity of Gemcitabine with AS1411.
Collapse
Affiliation(s)
- You Wu
- Graduate School of Pharmaceutical Science, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto, Japan
| | - Yusuke Kawamoto
- Graduate School of Pharmaceutical Science, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto, Japan
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, China.
| | - Yuki Takahashi
- Graduate School of Pharmaceutical Science, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto, Japan
| | - Yuriko Higuchi
- Graduate School of Pharmaceutical Science, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto, Japan.
| | - Yoshinobu Takakura
- Graduate School of Pharmaceutical Science, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto, Japan.
| |
Collapse
|
2
|
Li C, Zhang J, Yao X, Huang Y, Zhang Y, Yang W. Red blood cell membrane-camouflaged nanocarriers for the delivery of piperlongumine to treat triple-negative breast cancer. Biomed Mater 2025; 20:035034. [PMID: 40328285 DOI: 10.1088/1748-605x/add4da] [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: 12/11/2024] [Accepted: 05/06/2025] [Indexed: 05/08/2025]
Abstract
The application of the conventional drugs for triple-negative breast cancer (TNBC) treatment in chemotherapy is limited due to their intrinsic drawbacks such as short drug half-life, lack of tumor selectivity and systemic toxicity. Herein, an effective nanoparticle drug delivery system (NDDS) of red blood cell (RBC) membrane-camouflaged piperlongumine (PL)-loaded iron oxide (Fe3O4) magnetic nanoparticles (Fe3O4-PL@RBC) was rationally designed as an effective drug delivery platform forin vivoTNBC treatment. The Fe3O4-PL@RBC showed considerable cytotoxicity against MDA-MB-231 cells, inducing intracellular accumulation of reactive oxygen species, mitochondrial dysfunction and apoptosis. Furthermore, transcriptomic analyses and western blotting analysis demonstrated that the Fe3O4-PL@RBC induced apoptosis through the inhibition of PI3K/AKT/mTOR pathway and downregulation of Bcl-2 protein. In MDA-MB-231 tumor models, the RBC membrane coating in Fe3O4-PL@RBC effectively prolonged the circulation time and sufficient enrichment at the tumor sites. And the Fe3O4-PL@RBC significantly inhibited tumor growth with good biosafety. This study provides guidance for the rational design of effective Fe3O4-based NDDS for TNBC treatment.
Collapse
Affiliation(s)
- Chenxi Li
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Jiaxin Zhang
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Xianxian Yao
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Yuxin Huang
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Yichen Zhang
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers and Department of Macromolecular Science, Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China
| |
Collapse
|
3
|
Chatterjee O, Kaur GA, Shukla N, Balayan S, Singh PK, Chatterjee S, Tiwari A. Multifaceted arsenal in SELEX nanomedicine. Adv Colloid Interface Sci 2025; 342:103540. [PMID: 40344950 DOI: 10.1016/j.cis.2025.103540] [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: 02/06/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
Aptamers, short oligonucleotide sequences that bind specifically to cellular proteins and receptors, are emerging as versatile tools in molecular nanomedicine. Unlike passive tumor targeting via the enhanced permeability and retention (EPR) effect, aptamers enable precise drug delivery, enhancing therapeutic efficacy while minimizing side effects. Developed through the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) process, aptamers offer compact size, robust structure, chemical versatility, and cost-effective synthesis. They serve as effective delivery vehicles for therapeutic molecules, including miRNA, siRNA, and small-molecule drugs, and function as antibody-like ligands for applications in cancer, diabetes, and autoimmune disorders. Since the approval of Macugen, the first aptamer targeting VEGF, aptamers have also shown promise as diagnostic sensors and theranostic agents. This review explores SELEX-derived aptamers in nanomedicine, focusing on their therapeutic and diagnostic roles, particularly in precision cancer therapies. It also addresses challenges such as degradation and clinical translation alongside prospects in vaccines, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Oishika Chatterjee
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden; Department of Biological Sciences, Bose Institute Unified Academic Campus EN 80, Sector 5, Bidhan Nagar (Salt Lake City) Kolkata 700 091, WB, India
| | - Gun Anit Kaur
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden
| | - Nutan Shukla
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden
| | - Sapna Balayan
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden
| | - Pravin Kumar Singh
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden
| | - Subhrangsu Chatterjee
- Department of Biological Sciences, Bose Institute Unified Academic Campus EN 80, Sector 5, Bidhan Nagar (Salt Lake City) Kolkata 700 091, WB, India.
| | - Ashutosh Tiwari
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 590 53, Sweden.
| |
Collapse
|
4
|
Zhao D, Han Y, Chen Y, He Z, Xia Q, Ji DK, Tan W. Molecular Engineering of Glycoaptamer Dual-Target Radionuclide Probes for PET/CT Imaging of Triple-Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:25645-25653. [PMID: 40239107 DOI: 10.1021/acsami.5c00285] [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: 04/18/2025]
Abstract
Triple-negative breast cancer (TNBC) is one of the most malignant cancer types, characterized by a lack of efficient diagnostic and treatment methods in clinical practice. The development of effective targeted diagnosis and treatment for TNBC has become an important research focus. Here, we report the first proof-of-concept evidence of a glycoaptamer dual-target radionuclide probe (GDRP) for positron emission tomography/computed tomography (PET/CT) imaging of TNBC. The GDRP was created through precise molecular engineering of the aptamer AS1411, combined with three mannose ligands. The GDRP exhibited significantly increased serum stability and a high affinity for TNBC cells through a dual targeting mode. The advantages of our developed GDRP were further demonstrated by its excellent in vivo PET/CT dynamic imaging performance, featuring a long imaging window and high spatiotemporal resolution. This flexible method allows for the preparation of glycosylated functional nucleic acid molecular probes with high serum stability and tumor specificity. We anticipate that this PET/CT molecular probe will expand the development of glycoaptamer-based radioactive drugs and provides molecular tools for the clinical diagnosis of TNBC.
Collapse
Affiliation(s)
- Deyi Zhao
- School of Life Sciences, Shanghai University, Shanghai 200444, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yongqi Han
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yamei Chen
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zhenyang He
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qian Xia
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ding-Kun Ji
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Weihong Tan
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), The Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
5
|
Yang J, Wang L, Wen Y, Guo R, Huo Y, Zhao H, Li L, Yan J, Liu G. PolyA-Bridged Capture Probe Architecture Enables High-Efficiency DNA Hybridization for Multiplex Biosensing Applications. Anal Chem 2025; 97:9066-9075. [PMID: 40248894 DOI: 10.1021/acs.analchem.5c01752] [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: 04/19/2025]
Abstract
DNA target-probe hybridization is a critical recognition and combination process for establishing high-performance biosensors. However, in conventional self-assembly strategies, surface-anchored capture probes exhibit heterogeneous molecular conformations and limit the kinetics of DNA hybridization at the interface. As a result, the response speed and practicability of electrochemical biosensors are quite limited, especially in real samples. Interfacial regulation of the molecular conformation using artificial DNA nanostructures has been widely recognized as a promising strategy to improve the accessibility and activity of capture probes. This study introduces a significantly simplified molecular regulatory structure on the surface of the gold electrode consisting of a probe-polyA-probe (PAP) sequence and a capture probe (CP). The PAP sequence has a central polyA fragment anchoring to the gold electrode and two flanking probes for hybridization with the two ends of the capture probe, forming a bridged CP (BCP). Upon dual-terminal hybridization with PAP, the capture probe underwent structural linearization through opposing directional extension, thereby markedly enhancing the steric accessibility and subsequent hybridization efficiency. By establishing a BCP biosensor, we achieved rapid and sensitive detection of DNA hybridization from 1 fM to 1 nM. More importantly, the platform demonstrated valuable versatility in the construction of both a gap hybridization biosensor for microRNA and a DNAzyme biosensor for Pb2+. The BCP biosensor exhibited exceptional biorecognition capability, achieving rapid DNA hybridization kinetics in only 3 min and a remarkable hybridization efficiency of 95.56%. Based on its high sensitivity, operational simplicity, and broad applicability, our BCP biosensor has shown an avenue for the development of novel electrochemical biosensors for molecular diagnostics and environmental monitoring.
Collapse
Affiliation(s)
- Jiaqi Yang
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture; Shanghai Engineering Research Center of Aquatic-Product Process & Preservation; College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Lele Wang
- Key Laboratory of Bioanalysis and Metrology for State Market Regulation, Shanghai Institute of Measurement and Testing Technology, Shanghai 201203, China
| | - Yanli Wen
- Key Laboratory of Bioanalysis and Metrology for State Market Regulation, Shanghai Institute of Measurement and Testing Technology, Shanghai 201203, China
| | - Ruiyan Guo
- Key Laboratory of Bioanalysis and Metrology for State Market Regulation, Shanghai Institute of Measurement and Testing Technology, Shanghai 201203, China
| | - Yinbo Huo
- Key Laboratory of Bioanalysis and Metrology for State Market Regulation, Shanghai Institute of Measurement and Testing Technology, Shanghai 201203, China
| | - Haoran Zhao
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture; Shanghai Engineering Research Center of Aquatic-Product Process & Preservation; College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Lanying Li
- Key Laboratory of Bioanalysis and Metrology for State Market Regulation, Shanghai Institute of Measurement and Testing Technology, Shanghai 201203, China
| | - Juan Yan
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture; Shanghai Engineering Research Center of Aquatic-Product Process & Preservation; College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Gang Liu
- Key Laboratory of Bioanalysis and Metrology for State Market Regulation, Shanghai Institute of Measurement and Testing Technology, Shanghai 201203, China
| |
Collapse
|
6
|
Li L, Wei P, Kong T, Yuan B, Fu P, Li Y, Wang Y, Zheng J, Wang K. Framework nucleic acid-programmed aptamer-paclitaxel conjugates as targeted therapeutics for triple-negative breast cancer. NANOSCALE HORIZONS 2025; 10:873-884. [PMID: 40078065 DOI: 10.1039/d4nh00652f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Triple-negative breast cancer (TNBC) is highly invasive with a poor prognosis, and chemotherapy remains the clinical treatment of choice. Paclitaxel is a commonly used first-line chemotherapy drug, but its untargeted distribution poses clinical challenges. Inspired by antibody-drug conjugates, we develop a precisely structured framework nucleic acid-programmed aptamer-paclitaxel conjugate (FAPC) with chemically well-defined paclitaxel loading dosing, enabling the regulation of receptor-aptamer affinity to facilitate tumor-targeted chemotherapy. Utilizing framework nucleic acids as a precise addressing scaffold, we organize the AS1411 aptamer with accurate intermolecular spacing and find that an inter-aptamer spacing of 19.04 nm could enhance the affinity of the FAPC for tumor cells. Then, the multifunctional FAPC can disrupt actin reorganization to achieve cytotoxicity in tumor cells. Furthermore, the AS1411-specifically modified FAPC further enhances the structure-dependent selective accumulation of drugs at tumor sites in a human xenograft model of triple-negative breast cancer, subsequently leading to significantly improved antitumor efficacy and reduced toxicity. The FAPC provides a precisely programmable platform for efficient targeted delivery of chemotherapeutic agents to malignancies.
Collapse
Affiliation(s)
- Lin Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
| | - Pengyao Wei
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
| | - Tong Kong
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
| | - Bo Yuan
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
| | - Pan Fu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia
- St. George and Sutherland Clinical Campuses School of Clinical Medicine UNSW Sydney Kensington, NSW 2052, Australia
| | - Yuhui Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jianping Zheng
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Kaizhe Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering of Chinese Academy of Sciences, Ningbo, 315300, P. R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
7
|
Yang Y, Yang C, Deng K, Xiao Y, Liu X, Du Z. Nucleic Acid Drugs in Radiotherapy. Chembiochem 2025; 26:e202400854. [PMID: 39903093 DOI: 10.1002/cbic.202400854] [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: 10/15/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/06/2025]
Abstract
Radiotherapy remains a cornerstone of cancer treatment, using high-energy radiation to induce DNA damage in tumor cells, leading to cell death. However, its efficacy is often hindered by challenges such as radiation resistance and side effects. As a powerful class of functional molecules, nucleic acid drugs (NADs) present a promising solution to these limitations. Engineered to target key pathways like DNA repair and tumor hypoxia, NADs can enhance radiotherapy sensitivity. NADs can also serve as delivery vehicles for radiotherapy agents such as radionuclides, improving targeting accuracy and minimizing side effects. This review explores the role of NADs in optimizing radiotherapy, highlighting their mechanisms, clinical applications, and synergies with radiotherapy, ultimately offering a promising strategy for improving patient outcomes in cancer therapy.
Collapse
Affiliation(s)
- Yuying Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Cai Yang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Kai Deng
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Yating Xiao
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, Universities and Colleges Admissions Service (UCAS), Hangzhou, 310024, China
| | - Xiangsheng Liu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Zhen Du
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
8
|
de Souza MM, Gini ALR, Moura JA, Scarim CB, Chin CM, dos Santos JL. Prodrug Approach as a Strategy to Enhance Drug Permeability. Pharmaceuticals (Basel) 2025; 18:297. [PMID: 40143076 PMCID: PMC11946379 DOI: 10.3390/ph18030297] [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: 01/06/2025] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 03/28/2025] Open
Abstract
Absorption and permeability are critical physicochemical parameters that must be balanced to achieve optimal drug uptake. These key factors are closely linked to the maximum absorbable dose required to provide appropriate plasma levels of drugs. Among the various strategies employed to enhance drug solubility and permeability, prodrug design stands out as a highly effective and versatile approach for improving physicochemical properties and enabling the optimization of biopharmaceutical and pharmacokinetic parameters while mitigating adverse effects. Prodrugs are compounds with reduced or no activity that, through bio-reversible chemical or enzymatic processes, release an active parental drug. The application of this technology has led to significant advancements in drug optimization during the design phase, and it offers broad potential for further development. Notably, approximately 13% of the drugs approved by the U.S. Food and Drug Administration (FDA) between 2012 and 2022 were prodrugs. In this review article, we will explore the application of prodrug strategies to enhance permeability, describing examples of market drugs. We also describe the use of the prodrug approach to optimize PROteolysis TArgeting Chimeras (PROTACs) permeability by using conjugation technologies. We will highlight some new technologies in prodrugs to enrich permeability properties, contributing to developing new effective and safe prodrugs.
Collapse
Affiliation(s)
- Mateus Mello de Souza
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
| | - Ana Luísa Rodriguez Gini
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
| | - Jhonnathan Alves Moura
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-900, SP, Brazil;
| | - Cauê Benito Scarim
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
| | - Chung Man Chin
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
- Union of the Colleges of the Great Lakes (UNILAGO), School of Medicine, Advanced Research Center in Medicine (CEPAM), Sao Jose do Rio Preto 15030-070, SP, Brazil
| | - Jean Leandro dos Santos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil; (M.M.d.S.); (A.L.R.G.); (C.B.S.); (C.M.C.)
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-900, SP, Brazil;
| |
Collapse
|
9
|
Li S, Sah DK, Arjunan A, Ameer MY, Lee B, Jung YD. Triptolide suppresses IL-1β-induced expression of interleukin-8 by inhibiting ROS-Mediated ERK, AP-1, and NF-κB molecules in human gastric cancer AGS cells. Front Oncol 2025; 14:1498213. [PMID: 39950099 PMCID: PMC11821500 DOI: 10.3389/fonc.2024.1498213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/26/2024] [Indexed: 02/16/2025] Open
Abstract
Triptolide, the major component of Chinese herbal medicine Tripterygium wilfordii Hook F, possesses potent anticancer and anti-inflammatory effects. IL-8, a proinflammatory cytokine, is associated with cancer cell proliferation and angiogenesis. Here, we found that Triptolide has an inhibitory effect on IL-1β-induced IL-8 expression in human gastric cancer cells, via the suppression of reactive oxygen species (ROS) production, AP-1, and NF-κB activation, which in turn affects human endothelial cell angiogenetic activity in tumor microenvironments. Human gastric AGS cells were treated with IL-1β (10 ng/mL) and Triptolide (0-20 nM), and the ROS generation, ERK, AP-1, and NF-κB signaling were all investigated. These results demonstrate that Triptolide inhibits the IL-1β-induced IL-8 expression in gastric cancer cells by inhibiting ROS production and angiogenesis, via the dose-dependent attenuation of ERK, AP-1, and NF-κB activation. In this study, we showed that Triptolid inhibits ROS/ERK-mediated AP-1 and ROS-mediated NF-κB axes potentially leading to an improved treatment outcome for gastric cancer and its associated tumor microenvironment.
Collapse
Affiliation(s)
- Shinan Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Shanxi Medical University, Taiyuan, China
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Dhiraj Kumar Sah
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Archana Arjunan
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Mohamed Yazeer Ameer
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Bora Lee
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Young-Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Republic of Korea
| |
Collapse
|
10
|
Zheng X, Wang Y, Duan H, Hou J, He S. A new NCL-targeting aptamer-drug conjugate as a promising therapy against esophageal cancer. J Nanobiotechnology 2025; 23:52. [PMID: 39875991 PMCID: PMC11773979 DOI: 10.1186/s12951-025-03127-1] [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: 10/29/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025] Open
Abstract
Esophageal cancer (EC) is one of the most common highly malignant tumors of the digestive system, with a poor prognosis under current treatment regimens. Nucleolin (NCL) is overexpressed in many tumors, and drugs specifically targeting NCL may offer a promising strategy for treating esophageal cancer. Here, we designed and prepared a novel aptamer-conjugated drug targeting NCL by AS1411 aptamer-human serum albumin (HSA)-the apoprotein of lidamycin (LDP)-active enediyne chromophore (AE), in order to achieve targeted treatment of esophageal cancer. The experimental results revealed that AS1411-HSA-LDP effectively binds to esophageal cancer cells and could be efficiently internalized by esophageal cancer cells. In the KYSE520 xenograft tumor nude mouse model, AS1411-HSA-LDP could be targeted and enriched in the tumor location for a long time. AS1411-HSA-LDP-AE exhibited a strong cell-killing activity in esophageal cancer cells, inhibited cell migration and invasion, and induced cell apoptosis. The animal studies confirmed that AS1411-HSA-LDP-AE exhibited a strong anti-tumor effect. These findings suggested that the novel NCL-targeting aptamer-drug conjugate constructed based on lidamycin exhibited a strong anti-tumor effect, providing a promising strategy for the targeted treatment of esophageal cancer.
Collapse
Affiliation(s)
- Xue Zheng
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Ying Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Huaiyu Duan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Junqi Hou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Shiming He
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
11
|
Li J, Wang J, Chen Z. Emerging role of exosomes in cancer therapy: progress and challenges. Mol Cancer 2025; 24:13. [PMID: 39806451 PMCID: PMC11727182 DOI: 10.1186/s12943-024-02215-4] [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: 11/05/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025] Open
Abstract
This review highlights recent progress in exosome-based drug delivery for cancer therapy, covering exosome biogenesis, cargo selection mechanisms, and their application across multiple cancer types. As small extracellular vesicles, exosomes exhibit high biocompatibility and low immunogenicity, making them ideal drug delivery vehicles capable of efficiently targeting cancer cells, minimizing off-target damage and side effects. This review aims to explore the potential of exosomes in cancer therapy, with a focus on applications in chemotherapy, gene therapy, and immunomodulation. Additionally, challenges related to exosome production and standardization are analyzed, highlighting the importance of addressing these issues for their clinical application. In conclusion, exosome-based drug delivery systems offer promising potential for future cancer therapies. Further research should aim to enhance production efficiency and facilitate clinical translation, paving the way for innovative cancer treatment strategies.
Collapse
Affiliation(s)
- Jiale Li
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China
| | - Jiachong Wang
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China.
| | - Zigui Chen
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, China.
| |
Collapse
|
12
|
Cheng J, Dong G, Wang W, Sheng C. Precise Modulation of Protein Degradation by Smart PROTACs. Chembiochem 2025; 26:e202400682. [PMID: 39367518 DOI: 10.1002/cbic.202400682] [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: 08/17/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/06/2024]
Abstract
Proteolysis-targeting chimera (PROTAC) has emerged as an attractive therapeutic modality in drug discovery. PROTACs are bifunctional molecules that effectively bridge proteins of interest (POIs) with E3 ubiquitin ligases, such that, the target proteins are tagged with ubiquitin and subsequently degraded via the proteasome. Despite significant progress in the field of targeted protein degradation (TPD), the application of conventional PROTAC degraders still faces significant challenges, including systemic toxicity induced by non-tissue-specific targeting. To address this issue, a variety of smart PROTACs that can be activated by specific stimuli, have been developed for achieving conditional and spatiotemporal modulation of protein levels. Here, on the basis of our contributions, we overview recent advances of smart PROTACs, including tumor microenvironment-, photo-, and X-ray radiation-responsive PROTACs, that enable controllable TPD. The design strategy, case studies, potential applications and challenges will be focused on.
Collapse
Affiliation(s)
- Junfei Cheng
- Nautical Medicine Experimental Teaching Demonstration Center of Educational Institutions, Faculty of Naval Medicine, Second Military Medical University (Naval Medical University), Shanghai, 200433, People's Republic of China
| | - Guoqiang Dong
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai, 200433, People's Republic of China
| | - Wei Wang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai, 200433, People's Republic of China
| | - Chunquan Sheng
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Second Military Medical University (Naval Medical University), Shanghai, 200433, People's Republic of China
| |
Collapse
|
13
|
Zheng X, Huang Z, Zhang Q, Li G, Song M, Peng R. Aptamer-functionalized nucleic acid nanotechnology for biosensing, bioimaging and cancer therapy. NANOSCALE 2025; 17:687-704. [PMID: 39585179 DOI: 10.1039/d4nr04360j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Nucleic acids have enabled the fabrication of self-assemblies and dynamic operations. Among different functional nucleic acids, aptamers can specifically bind to a wide range of targets, including proteins, viral antigens, living cells and even tissues, and have thus emerged as molecular recognition tools in molecular medicine. Hence, aptamer-functionalized nucleic acid nanotechnology offers applications of biosensing, bioimaging, and cancer therapy. In this review, after a brief overview of nucleic acid nanotechnology, we focus on the integration of aptamers with nucleic acid nanotechnology, including self-assembly constructions and dynamic molecular manipulations. The emerging applications in molecular medicine are subsequently reviewed with aptamer-based self-assemblies and aptamer-involved dynamic molecular manipulation. For convenience, applications are broadly categorized into biosensing, bioimaging, and cancer therapy. Finally, challenges and potential development of nucleic acid nanotechnology are discussed.
Collapse
Affiliation(s)
- Xiaofang Zheng
- Zhejiang Cancer Hospital, Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China.
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing 400030, P. R. China
| | - Zhiyong Huang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, P. R. China
| | - Qiang Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, P. R. China
| | - Guoli Li
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing 400030, P. R. China
| | - Minghui Song
- Zhejiang Cancer Hospital, Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China.
| | - Ruizi Peng
- Zhejiang Cancer Hospital, Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P. R. China.
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, P. R. China
| |
Collapse
|
14
|
Han Y, Zhang R, Bao H, Yang M, Gao Y, Gao X, Wang R, Tan W, Ji D. Molecular Programming Design of Glyconucleic Acid Aptamer with High Stability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408168. [PMID: 39630080 PMCID: PMC11775523 DOI: 10.1002/advs.202408168] [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: 07/17/2024] [Revised: 11/15/2024] [Indexed: 01/30/2025]
Abstract
Functional nucleic acids (FNAs), possessing specific biological functions beyond their informational roles, have gained widespread attention in disease therapeutics. However, their clinical application is severely limited by their low serum stability in complex physiological environments. In this work, a precise molecular programming strategy is explored to prepare glyconucleic acid aptamers (GNAAs) with high serum stability. Four glyconucleic acid modules compatible with commercial solid-phase synthesis are designed and synthesized. Through precise molecular design, the accurate modification of four different carbohydrate ligands at specific sites of DNA aptamers is achieved. It is demonstrated that glycosylation modification can significantly increase DNA aptamers' serum stability while maintaining their structures and high affinity. The stabilization effect is superior to that of currently commonly used commercial chemical modifications. Moreover, it is confirmed that this approach displays insignificant effects on the DNA aptamers' tumor-targeting ability and metabolism in vivo. This method offers a simple, economical, and efficient strategy for precise glycosylation modification of nucleic acids. This allows to prepare glycosyl functional nucleic acids with high serum stability, which can expand the application scope of functional nucleic acids and promote the practical transformation of functional nucleic acids.
Collapse
Affiliation(s)
- Yongqi Han
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
- College of Chemistry and Materials ScienceShanghai Normal UniversityShanghai200234China
| | - Rongjun Zhang
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
| | - Hong‐Liang Bao
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
| | - Mei Yang
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
| | - Yuan Gao
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
| | - Xiaobo Gao
- Department of Anatomy and PhysiologyShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Ruowen Wang
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
- Zhejiang Cancer HospitalHangzhou Institute of Medicine (HIM)The Chinese Academy of SciencesHangzhouZhejiang310022China
- Molecular Science and Biomedicine Laboratory (MBL)State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Chemistry and Chemical EngineeringCollege of BiologyAptamer Engineering Center of Hunan ProvinceHunan University ChangshaHunan410082China
| | - Ding‐Kun Ji
- Institute of Molecular Medicine (IMM)Renji HospitalState Key Laboratory of Oncogenes and Related GenesShanghai Jiao Tong University School of MedicineShanghai200240China
| |
Collapse
|
15
|
Xiong H, Song Z, Wang T, Huang K, Yu F, Sun W, Liu X, Liu L, Jiang H, Wang X. Photoswitchable dynamics and RNAi synergist with tailored interface and controlled release reprogramming tumor immunosuppressive niche. Biomaterials 2025; 312:122712. [PMID: 39098305 DOI: 10.1016/j.biomaterials.2024.122712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/29/2024] [Accepted: 07/25/2024] [Indexed: 08/06/2024]
Abstract
Immunosuppressive tumor microenvironment (ITM) severely limited the efficacy of immunotherapy against triple-negative breast cancer (TNBC). Herein, Apt-LPR, a light-activatable photodynamic therapy (PDT)/RNAi immune synergy-enhancer was constructed by co-loading miR-34a and photosensitizers in cationic liposomes (in phase III clinical trial). Interestingly, the introduction of tumor-specific aptamers creates a special "Liposome-Aptamer-Target" interface, where the aptamers are initially in a "lying down" state but transform to "standing up" after target binding. The interfacing mechanism was elaborately revealed by computational and practical experiments. This unique interface endowed Apt-LPR with neutralized surface potential of cationic liposomes to reduce non-specific cytotoxicity, enhanced DNase resistance to protect aptamers, and preserved target-binding ability for selective drug delivery. Upon near-infrared irradiation, the generated reactive oxygen species would oxidize unsaturated phospholipids to destabilize both liposomes and lysosomes, realizing stepwise lysosomal escape of miR-34a for tumor cell apoptosis and downregulation of PD-L1 to suppress immune escape. Together, tumor-associated antigens released from PDT-damaged mitochondria and endoplasmic reticulum could activate the suppressive immune cells to establish an "immune hot" milieu. The collaborative immune-enhancing strategy effectively aroused systemic antitumor immunity and inhibited primary and distal tumor progression as well as lung metastasis in 4T1 xenografted mouse models. The photo-controlled drug release and specific tumor-targeting capabilities of Apt-LPR were also visualized in MDA-MB-231 xenografted zebrafish models. Therefore, this photoswitchable PDT/RNAi immune stimulator offered a powerful approach to reprogramming ITM and reinforcing cancer immunotherapy efficacy.
Collapse
Affiliation(s)
- Hongjie Xiong
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Zhongquan Song
- Department of Respiratory Medicine, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, PR China
| | - Tingya Wang
- Department of Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, PR China
| | - Ke Huang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Fangfang Yu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Wenyu Sun
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Liu Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China.
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China.
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China.
| |
Collapse
|
16
|
Tang H, Zhang Y, Wu Y, Fu T, Cui C, Wang Z, Xie S, Wu Q, Tan W. The Emerging Era of Molecular Medicine. ACS NANO 2024; 18:30911-30918. [PMID: 39475564 DOI: 10.1021/acsnano.4c07969] [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: 11/13/2024]
Abstract
The era of molecular medicine arose as we began to diagnose and treat diseases based on understanding how genes, proteins, and cells work, providing optimal therapeutic care through molecular profiling. Central to molecular medicine is molecular recognition, which is underpinned by techniques involving omics analysis, gene editing, and targeted agents. Recent advancements in these tools not only expand our understanding of biological processes but also aid in the development of diagnostic and treatment modalities at the molecular level, thus bridging the gap between medical research and clinical applications. This perspective traces the development of molecular tools, highlighting, along the way, their pivotal role in advancing molecular medicine for the global health of people.
Collapse
Affiliation(s)
- Heming Tang
- Department of Urology, Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ying Zhang
- Department of Geriatric Endocrinology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yanyuan Wu
- Department of Urology, Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ting Fu
- Department of Urology, Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Cheng Cui
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Zongping Wang
- Department of Urology, Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Sitao Xie
- Department of Urology, Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Qin Wu
- Department of Urology, Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Nucleic Acids, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
17
|
Li M, Li J, Tang Q, Zhu Y. Potential antitumor activity of triptolide and its derivatives: Focused on gynecological and breast cancers. Biomed Pharmacother 2024; 180:117581. [PMID: 39427548 DOI: 10.1016/j.biopha.2024.117581] [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: 08/06/2024] [Revised: 10/01/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024] Open
Abstract
Cancer remains one of the greatest global health concerns. This is especially true for gynecological cancers, which include cervical, ovarian, and endometrial cancers, and breast cancer. Natural products used for cancer treatment offer some unique advantages. Triptolide (TPL) is a biologically active terpenoid extracted from Tripterygium wilfordii, which exhibits anti-inflammatory, immunosuppressive, antitumor, and other pharmacological activities. However, clinical applications of TPL are restricted because of poor water solubility and severe cytotoxicity; to overcome these limitations, various TPL derivatives and drug delivery systems, especially nanocarriers, have been used. Furthermore, various preclinical and clinical studies have demonstrated that TPL and its derivatives exhibit excellent antitumor effects by targeting proteins involved in multiple signaling pathways. Here, we review the progress regarding novel drug delivery systems, antitumor activities, and molecular mechanisms of action of TPL and its derivatives against gynecological and breast cancers. TPL and its derivatives inhibit tumor growth, suppress tumor metastasis, and enhance the drug sensitization of resistant cancers. In addition, TPL and its derivatives exert synergistic antitumor effects against gynecological and breast cancers when combined with existing antitumor drugs, such as carboplatin, cisplatin, and PI3K inhibitors. Moreover, we highlight the clinical potential of TPL analogs against cancer from bench to bedside and their prospects for future applications in gynecologic and breast cancers.
Collapse
Affiliation(s)
- Mengjie Li
- College of Pharmacy, Qinghai University for Nationalities, Xining, China; Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jiamiao Li
- Department of Pharmacy, The Affilliated Chengdu 363 Hospital of Southwest Medical University, Chengdu, China
| | - Qing Tang
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yongxia Zhu
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
18
|
Chen X, Li D, Su Q, Ling X, Yang Y, Liu Y, Zhu X, He A, Ding S, Xu R, Liu Z, Long X, Zhang J, Yang Z, Qi Y, Wu H. SENP3 mediates the deSUMOylation and degradation of YAP1 to regulate the progression of triple-negative breast cancer. J Biol Chem 2024; 300:107764. [PMID: 39270822 PMCID: PMC11490879 DOI: 10.1016/j.jbc.2024.107764] [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: 03/17/2024] [Revised: 07/23/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a prevalent malignancy in women, casting a formidable shadow on their well-being. Positioned within the nucleolus, SUMO-specific protease 3 (SENP3) assumes a pivotal role in the realms of development and tumorigenesis. However, the participation of SENP3 in TNBC remains a mystery. Here, we elucidate that SENP3 exerts inhibitory effects on migration and invasion capacities, as well as on the stem cell-like phenotype, within TNBC cells. Further experiments showed that YAP1 is the downstream target of SENP3, and SENP3 regulates tumorigenesis in a YAP1-dependent manner. YAP1 is found to be SUMOylated and SENP3 deconjugates SUMOylated YAP1 and promotes degradation mediated by the ubiquitin-proteasome system. More importantly, YAP1 with a mutation at the SUMOylation site impedes the capacity of WT YAP1 in TNBC tumorigenesis. Taken together, our findings firmly establish the pivotal role of SENP3 in the modulation of YAP1 deSUMOylation, unveiling novel mechanistic insight into the important role of SENP3 in the regulation of TNBC tumorigenesis in a YAP1-dependent manner.
Collapse
Affiliation(s)
- Xu Chen
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Danqing Li
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Qi Su
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Xing Ling
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Yanyan Yang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Yuhang Liu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Xinjie Zhu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Anqi He
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Siyu Ding
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Runxiao Xu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Zhaoxia Liu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Xiaojun Long
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Jinping Zhang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhihui Yang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yitao Qi
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China.
| | - Hongmei Wu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China.
| |
Collapse
|
19
|
Wang L, Yin H, Jiang J, Li Q, Gao C, Li W, Zhang B, Xin Y, Li H, Zhao M, Lu Q. A rationally designed CD19 monoclonal antibody-triptolide conjugate for the treatment of systemic lupus erythematosus. Acta Pharm Sin B 2024; 14:4560-4576. [PMID: 39525579 PMCID: PMC11544386 DOI: 10.1016/j.apsb.2024.06.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 11/16/2024] Open
Abstract
Tripterygium wilfordii Hook F (TWHF) is a traditional Chinese medicine widely used in the treatment of systemic lupus erythematosus (SLE), with triptolide (TP) as its main active ingredient. However, its side effects also induced by TP, especially hepatotoxicity and reproductive toxicity, largely limit its application in a subset of patients. Monoclonal antibodies (mAbs) developed for the treatment of SLE that deplete B cells by targeting B cell-expressing antigens, such as CD19, have failed in clinical trials, partly due to their poor efficacy in consuming B cells. Here, we report the development of a rationally designed antibody‒drug conjugate (ADC), CD19 mAb-TP conjugate, to alleviate the side effects of TWHF and simultaneously improve the therapeutic efficacy of CD19 mAb. The CD19 mAb-TP conjugate, which was named ADC-TP, selectively depleted B cell subsets both in vitro and in vivo and effectively alleviated disease symptoms in mouse lupus models with enhanced therapeutic efficacy than CD19 mAb and fewer side effects than TP. Our present study proposes a CD19 mAb‒TP conjugate strategy to mitigate the toxicity of TWHF while also enhancing the therapeutical efficacy of CD19 mAbs for the treatment of SLE, providing a feasible method for improving the current agents used for treating SLE.
Collapse
Affiliation(s)
- Lai Wang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China
| | - Haoyuan Yin
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China
| | - Jiao Jiang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China
| | - Qilin Li
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China
- Hunan Key Laboratory of Medical Epigenomics, the Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Changxing Gao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China
| | - Wenrui Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China
| | - Bo Zhang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China
| | - Yue Xin
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China
| | - Hongyang Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing 210042, China
| | - Ming Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China
| |
Collapse
|
20
|
Wang X, Seah HL, Zhang XL, Zhuang Z, Liu XW. Fluorescent Self-Assembled Complexes Based on Glyco-Functionalized G-Quadruplexes as a Targeted Delivery Platform. ACS APPLIED MATERIALS & INTERFACES 2024; 16:50229-50237. [PMID: 39264898 DOI: 10.1021/acsami.4c08079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
Targeted delivery systems combined with the stimuli-responsive release of drug molecules hold noteworthy promise for precision medicine, enabling treatments with enhanced effectiveness and reduced adverse effects. An ideal drug delivery platform with versatile targeting moieties, the capability of combinational payloads, and simple preparation is highly desirable. Herein, we developed pH-sensitive fluorescent self-assembled complexes (SACs) of a galactose-functionalized G-quadruplex (G4) and a coumarin carboxamidine derivative as a targeted delivery platform through the nanoprecipitation method. These SACs selectively targeted hepatocellular carcinoma (HepG2) cells in fluorescence imaging after a short incubation and exerted specific anticancer effects in an appropriate dose range. Co-delivery of 1 μM prodrug floxuridine oligomers and 16 μg/mL SACs (minimal hemolytic effect) significantly reduced the cytotoxicity of the nucleoside anticancer drug on normal cells (NIH/3T3), kept up to 70% alive after 72-h incubation, and improved anticancer efficacy compared to SACs alone. This strategy can be extended to ratiometric multidrug delivery through self-assembly for targeted combinational therapy.
Collapse
Affiliation(s)
- Xian Wang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Hui Ling Seah
- School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Xiao-Lin Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Zeyan Zhuang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Xue-Wei Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| |
Collapse
|
21
|
Krishna A, Raj G, P S, Mohan G, Aliyas BB, Perumal D, Varghese R. Esterase-Responsive Floxuridine-Tethered Multifunctional Nanoparticles for Targeted Cancer Therapy. ACS APPLIED BIO MATERIALS 2024; 7:6276-6285. [PMID: 39215722 DOI: 10.1021/acsabm.4c00886] [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] [Indexed: 09/04/2024]
Abstract
Floxuridine is a potential clinical anticancer drug for the treatment of various cancers. However, floxuridine typically causes unfavorable side effects due to its very poor tumor selectivity, and, hence, there is a high demand for the development of novel approaches that permit the targeted delivery of floxuridine into cancerous cells. Herein, the design and synthesis of an esterase-responsive multifunctional nanoformulation for the targeted delivery of floxuridine in esterase-overexpressed cancer cells is reported. Photopolymerization of floxuridine-tethered lipoic acid results in the formation of amphiphilic floxuridine-tethered poly(disulfide). Self-assembly of the amphiphilic polymer results in the formation of nanoparticles with floxuridine decorated on the surfaces of the particles. Integration of aptamer DNA for nucleolin onto the surface of the nanoparticle is demonstrated by exploring the base-pairing interaction of floxuridine with adenine. Targeted internalization of the aptamer-decorated nanoparticle into nucleolin-expressed cancer cells is demonstrated. Esterase triggered cleavage of the ester bond connecting floxuridine with the polymer backbone, and the subsequent targeted delivery of floxuridine into cancer cells is also shown. Excellent therapeutic efficacy is observed both in vitro and also in the 3D tumor spheroid model. This noncovalent strategy provides a simple yet effective strategy for the targeted delivery of floxuridine into cancer cells in a less laborious fashion.
Collapse
Affiliation(s)
- Anusree Krishna
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram 695551, Kerala India
| | - Gowtham Raj
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram 695551, Kerala India
| | - Sandhya P
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram 695551, Kerala India
| | - Ganga Mohan
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram 695551, Kerala India
| | - Basil B Aliyas
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram 695551, Kerala India
| | - Devanathan Perumal
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram 695551, Kerala India
| | - Reji Varghese
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram 695551, Kerala India
| |
Collapse
|
22
|
Li C, Shi K, Zhao S, Liu J, Zhai Q, Hou X, Xu J, Wang X, Liu J, Wu X, Fan W. Natural-source payloads used in the conjugated drugs architecture for cancer therapy: Recent advances and future directions. Pharmacol Res 2024; 207:107341. [PMID: 39134188 DOI: 10.1016/j.phrs.2024.107341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024]
Abstract
Drug conjugates are obtained from tumor-located vectors connected to cytotoxic agents via linkers, which are designed to deliver hyper-toxic payloads directly to targeted cancer cells. These drug conjugates include antibody-drug conjugates (ADCs), peptide-drug conjugates (PDCs), small molecule-drug conjugates (SMDCs), nucleic acid aptamer-drug conjugates (ApDCs), and virus-like drug conjugate (VDCs), which show great therapeutic value in the clinic. Drug conjugates consist of a targeting carrier, a linker, and a payload. Payloads are key therapy components. Cytotoxic molecules and their derivatives derived from natural products are commonly used in the payload portion of conjugates. The ideal payload should have sufficient toxicity, stability, coupling sites, and the ability to be released under specific conditions to kill tumor cells. Microtubule protein inhibitors, DNA damage agents, and RNA inhibitors are common cytotoxic molecules. Among these conjugates, cytotoxic molecules of natural origin are summarized based on their mechanism of action, conformational relationships, and the discovery of new derivatives. This paper also mentions some cytotoxic molecules that have the potential to be payloads. It also summarizes the latest technologies and novel conjugates developed in recent years to overcome the shortcomings of ADCs, PDCs, SMDCs, ApDCs, and VDCs. In addition, this paper summarizes the clinical trials conducted on conjugates of these cytotoxic molecules over the last five years. It provides a reference for designing and developing safer and more efficient conjugates.
Collapse
Affiliation(s)
- Cuiping Li
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Kourong Shi
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Siyuan Zhao
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Juan Liu
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Qiaoli Zhai
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Xiaoli Hou
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Jie Xu
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Xinyu Wang
- Shanghai Wei Er Lab, Shanghai 201707, China.
| | - Jiahui Liu
- Fujian University of Traditional Chinese Medicine, Fuzhou 350108, China.
| | - Xin Wu
- Fujian University of Traditional Chinese Medicine, Fuzhou 350108, China; Shanghai Wei Er Lab, Shanghai 201707, China.
| | - Wei Fan
- Department of Pharmacy, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| |
Collapse
|
23
|
Zhu S, Gao H, Li W, He X, Jiang P, Xu F, Jin G, Guo H. Stimuli-Responsive Aptamer-Drug Conjugates for Targeted Drug Delivery and Controlled Drug Release. Adv Healthc Mater 2024; 13:e2401020. [PMID: 38742703 DOI: 10.1002/adhm.202401020] [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: 03/18/2024] [Revised: 04/21/2024] [Indexed: 05/16/2024]
Abstract
Chemotherapy is widely used for cancer therapy but with unsatisfied efficacy, mainly due to the inefficient delivery of anticancer agents. Among the critical "five steps" drug delivery process, internalization into tumor cells and intracellular drug release are two important steps for the overall therapeutic efficiency. Strategy based on active targeting or TME-responsive is developed individually to improve therapeutic efficiency, but with limited improvement. However, the combination of these two strategies could potentially augment the drug delivery efficiency and therapeutic efficiency, consequently. Therefore, this work constructs a library of stimuli-responsive aptamer-drug conjugates (srApDCs), as "dual-targeted" strategy for cancer treatment that enables targeted drug delivery and controlled drug release. Specifically, this work uses different stimuli-responsive linkers to conjugate a tumor-targeting aptamer (i.e., AS1411) with drugs, forming the library of srApDCs for targeted cancer treatment. This design hypothesis is validated by the experimental data, which indicated that the aptamer could selectively enhance uptake of the srApDCs and the linkers could be cleaved by pathological cues in the TME to release the drug payload, leading to a significant enhancement of therapeutic efficacy. These results underscore the potential of the approach, providing a promising methodology for cancer therapy.
Collapse
Affiliation(s)
- Shanshan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Huan Gao
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Wenyuan Li
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Xiaocong He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Panpan Jiang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Guorui Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Hui Guo
- First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
24
|
Raj G, Ghosh T, D S V, P H, Kumar DB, Prasad J, V B A, S M A, Varghese R. G 4-Hemin-loaded 2D nanosheets for combined and targeted chemo-photodynamic cancer therapy. NANOSCALE 2024; 16:16195-16203. [PMID: 39140185 DOI: 10.1039/d4nr01494d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Synergetic combination therapy is emerging as one of the most promising approaches for cancer treatment. Among the various therapeutic approaches, PDT has received particular attention due to its non-invasive nature. However, the therapeutic performance of PDT is severely affected by tumour hypoxia. Herein, we report a supramolecular strategy for the fabrication of a PDT-active 2D nanosheet loaded with a POD mimicking DNAzyme for the synergetic combination of PDT and CDT for targeted cancer therapy. Assembly of biotin-functionalized BODIPY (1) and cationic β-cyclodextrin (β-CD+) leads to the formation of a 1/β-CD+ nanosheet with positively charged β-CD+ on the surface of the sheet. The cationic face of the 1/β-CD+ sheet was then loaded with a POD-mimicking Hem-loaded G-quadruplex aptamer (Hem/DNA1) via electrostatic interactions (1/β-CD+/Hem/DNA1). Cellular internalization of the 1/β-CD+/Hem/DNA1 nanosheet occurs via a receptor-mediated endocytic pathway, which then undergoes lysosomal escape. Subsequently, Hem/DNA1 on the surface of 1/β-CD+/Hem/DNA1 reacts with endogenous H2O2via the Fenton pathway to produce ˙OH and O2. Moreover, under cellular conditions, Hem inside the 1/β-CD+/Hem/DNA1 nanosheet produces Fe2+, which then undergoes another Fenton reaction to produce ˙OH and O2. The Fe3+ generated after the Fenton reaction is then reduced in situ to Fe2+ by glutathione for the next Fenton cycle. At the same time, photoirradiation of the 1/β-CD+ nanosheet using a 635 nm laser produces 1O2via the PDT pathway by using endogenous O2. The most remarkable feature of the present nanoformulation is the cooperativity in its therapeutic action, wherein O2 produced during the CDT pathway was used by the 1/β-CD+ sheet for improving its PDT efficacy in the hypoxic tumor microenvironment. This work represents a unique combination of CDT and PDT for targeted cancer therapy, wherein the CDT action of the nanoagent enhances the PDT efficacy and we strongly believe that this approach would encourage researchers to design similar combination therapy for advancements in the treatment of cancer.
Collapse
Affiliation(s)
- Gowtham Raj
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| | - Tamraparni Ghosh
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| | - Vasudev D S
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| | - Harsha P
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| | - Devu B Kumar
- School of Biology, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India
| | - Justin Prasad
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| | - Athul V B
- School of Biology, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India
| | - Abhimanyu S M
- School of Biology, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India
| | - Reji Varghese
- School of Chemistry, Indian Institute of Science Education and Research (IISER) Thiruvananthapuram, Trivandrum-695551, Kerala, India.
| |
Collapse
|
25
|
Liu X, Zhao P, Du X, Hou J, Zhang G, Zhang W, Yang L, Chen Y. Let-7b-5p promotes triptolide-induced growth-inhibiting effects in glioma by targeting IGF1R. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5909-5925. [PMID: 38363352 DOI: 10.1007/s00210-024-02957-4] [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: 09/27/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024]
Abstract
Glioma is one of the most common malignancies of the central nervous system. The therapeutic effect has not been satisfactory despite advances in comprehensive treatment techniques. Our previous studies have found that triptolide inhibits glioma proliferation through the ROS/JNK pathway, but in-depth mechanisms need to be explored. Recent studies have confirmed that miRNAs may function as tumor suppressor genes or oncogenes and be involved in cancer development and progression. In this study, we found that let-7b-5p expression levels closely correlated with WHO grades and overall survival in patients in tumor glioma-CGGA-mRNAseq-325, and the upregulation of let-7b-5p can inhibit the proliferation and induce apoptosis of glioma cells. Functionally, upregulation of let-7b-5p increased the inhibitory effect on cell viability and colony formation caused by triptolide and promoted the apoptosis rate of triptolide-treated U251 cells. Conversely, downregulation of let-7b-5p had the opposite effect, indicating that let-7b-5p is a tumor suppressor miRNA in glioma cells. Moreover, target prediction, luciferase reporter assays and functional experiments revealed that IGF1R was a direct target of let-7b-5p. In addition, upregulation of IGF1R reversed the triptolide-regulated inhibition of cell viability but promoted glioma cell apoptosis and activated the ROS/JNK signaling pathway induced by triptolide. The results obtained in vivo experiments substantiated those from the in vitro experiments. In summary, the current study provides evidence that triptolide inhibits the growth of glioma cells by regulating the let-7b-5p-IGF1R-ROS/JNK axis in vitro and in vivo. These findings may provide new ideas and potential targets for molecularly targeted therapies for comprehensive glioma treatment.
Collapse
Affiliation(s)
- Xihong Liu
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, NO. 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Peiyuan Zhao
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, NO. 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Xiaodan Du
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, NO. 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Junlin Hou
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, NO. 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Guanghui Zhang
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Wenxian Zhang
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, NO. 156 Jinshui East Road, Zhengzhou, 450046, China
| | - Liping Yang
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, NO. 156 Jinshui East Road, Zhengzhou, 450046, China.
| | - Yulong Chen
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, NO. 156 Jinshui East Road, Zhengzhou, 450046, China.
| |
Collapse
|
26
|
Yang X, He L, Li X, Wang L, Bu T, Yun D, Lu X, Gao S, Huang Q, Li J, Zheng B, Yu J, Sun F. Triptolide exposure triggers testicular vacuolization injury by disrupting the Sertoli cell junction and cytoskeletal organization via the AKT/mTOR signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 279:116502. [PMID: 38788563 DOI: 10.1016/j.ecoenv.2024.116502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Despite the known reproductive toxicity induced by triptolide (TP) exposure, the regulatory mechanism underlying testicular vacuolization injury caused by TP remains largely obscure. METHODS Male mice were subjected to TP at doses of 15, 30, and 60 μg/kg for 35 consecutive days. Primary Sertoli cells were isolated from 20-day-old rat testes and exposed to TP at concentrations of 0, 40, 80, 160, 320, and 640 nM. A Biotin tracer assay was conducted to assess the integrity of the blood-testis barrier (BTB). Transepithelial electrical resistance (TER) assays were employed to investigate BTB function in primary Sertoli cells. Histological structures of the testes and epididymides were stained with hematoxylin and eosin (H&E). The expression and localization of relevant proteins or pathways were assessed through Western blotting or immunofluorescence staining. RESULTS TP exposure led to dose-dependent testicular injuries, characterized by a decreased organ coefficient, reduced sperm concentration, and the formation of vacuolization damage. Furthermore, TP exposure disrupted BTB integrity by reducing the expression levels of tight junction (TJ) proteins in the testes without affecting basal ectoplasmic specialization (basal ES) proteins. Through the TER assay, we identified that a TP concentration of 160 nM was optimal for elucidating BTB function in primary Sertoli cells, correlating with reductions in TJ protein expression. Moreover, TP exposure induced changes in the distribution of the BTB and cytoskeleton-associated proteins in primary Sertoli cells. By activating the AKT/mTOR signaling pathway, TP exposure disturbed the balance between mTORC1 and mTORC2, ultimately compromising BTB integrity in Sertoli cells. CONCLUSION This investigation sheds light on the impacts of TP exposure on testes, elucidating the mechanism by which TP exposure leads to testicular vacuolization injury and offering valuable insights into comprehending the toxic effects of TP exposure on testes.
Collapse
Affiliation(s)
- Xiwen Yang
- School of Basic Medical Science, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Lei He
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Xinyao Li
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Lingling Wang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Tiao Bu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Damin Yun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Xinran Lu
- School of Basic Medical Science, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| | - Sheng Gao
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Qiuru Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Jiaxin Li
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Bo Zheng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School of Nanjing Medical University, Suzhou 215002, China.
| | - Jun Yu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| | - Fei Sun
- School of Basic Medical Science, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan 750004, China; Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| |
Collapse
|
27
|
Zhao D, Li Z, Ji DK, Xia Q. Recent Progress of Multifunctional Molecular Probes for Triple-Negative Breast Cancer Theranostics. Pharmaceutics 2024; 16:803. [PMID: 38931924 PMCID: PMC11207493 DOI: 10.3390/pharmaceutics16060803] [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: 05/14/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Breast cancer (BC) poses a significant threat to women's health, with triple-negative breast cancer (TNBC) representing one of the most challenging and aggressive subtypes due to the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. Traditional TNBC treatments often encounter issues such as low drug efficiency, limited tumor enrichment, and substantial side effects. Therefore, it is crucial to explore novel diagnostic and treatment systems for TNBC. Multifunctional molecular probes (MMPs), which integrate target recognition as well as diagnostic and therapeutic functions, introduce advanced molecular tools for TNBC theranostics. Using an MMP system, molecular drugs can be precisely delivered to the tumor site through a targeted ligand. Real-time dynamic monitoring of drug release achieved using imaging technology allows for the evaluation of drug enrichment at the tumor site. This approach enables accurate drug release, thereby improving the therapeutic effect. Therefore, this review summarizes the recent advancements in MMPs for TNBC theranostics, encompassing the design and synthesis of MMPs as well as their applications in the field of TNBC theranostics.
Collapse
Affiliation(s)
- Deyi Zhao
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (D.Z.); (Z.L.)
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Zhe Li
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (D.Z.); (Z.L.)
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ding-Kun Ji
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Qian Xia
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
28
|
Fang Y, Wu Q, Wang F, Liu Y, Zhang H, Yang C, Zhu Z. Aptamer-RIBOTAC Strategy Enabling Tumor-Specific Targeted Degradation of MicroRNA for Precise Cancer Therapy. SMALL METHODS 2024:e2400349. [PMID: 38794853 DOI: 10.1002/smtd.202400349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/08/2024] [Indexed: 05/26/2024]
Abstract
MicroRNA (miRNA) molecules play crucial roles in a variety of diseases, making miRNA targeting a burgeoning field in medicinal chemistry. Ribonuclease targeting chimeras (RIBOTACs) present a compelling approach for RNA degradation. However, small molecule-based RIBOTAC requires an expensive and time-consuming screening process, and is difficult to directly target miRNA due to its short length lacking secondary structure. Antisense oligonucleotide (ASO)-based RIBOTAC is easy to design but with poor cell permeability. While both of them lack the specificity for tumor targeting. In this study, the first Aptamer-RIBOTAC (ARIBOTAC) chimera is designed based on ASO to achieve precise degradation of miRNA in a tumor cell-specific manner for precise cancer therapy. This chimera exhibits a remarkable ability to specifically identify and enter cancer cells, trigger localized activation of endogenous RNase L, and selectively cleave miRNAs that are complementary to ASO. The efficacy and universality of the ARIBOTAC strategy both in vitro and in vivo by degrading oncogenic miR-210-3p and miR-155-5p are validated. These findings underscore the potential of the ARIBOTAC strategy as a promising avenue for cancer therapy by precisely targeting cancer-associated miRNAs.
Collapse
Affiliation(s)
- Yuan Fang
- MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361000, China
| | - Qiuyue Wu
- MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361000, China
| | - Feiyu Wang
- MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361000, China
| | - Ye Liu
- MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361000, China
| | - Huimin Zhang
- Innovation Laboratory for Sciences, Technologies of Energy Materials of Fujian Province, Xiamen, 361000, China
| | - Chaoyong Yang
- MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361000, China
- Innovation Laboratory for Sciences, Technologies of Energy Materials of Fujian Province, Xiamen, 361000, China
| | - Zhi Zhu
- MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Key Laboratory for Chemical Biology of Fujian Province, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361000, China
| |
Collapse
|
29
|
Li Y, Zhang C, Fu T, Wang XQ, Tan W. Polyfluoroalkyl Tag Decoration Enables Significantly Enhanced Tumor Penetration Ability of a PTK7 Targeting Aptamer. Bioconjug Chem 2024; 35:674-681. [PMID: 38695582 DOI: 10.1021/acs.bioconjchem.4c00131] [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: 05/16/2024]
Abstract
Aptamers are widely used molecular recognition tools in targeted therapy, but their ability to effectively penetrate deep into solid tumors remains a significant challenge, leading to suboptimal treatment efficacy. Here, we developed a polyfluoroalkyl (PFA) decoration strategy to enhance aptamer recognition, cell internalization, and solid tumor penetration. Our results indicate that PFA with around 11 fluorine atoms significantly improves aptamer internalization both in vitro and in vivo settings. However, we also observed that the use of PFA tags containing 19 and 23 fluorine atoms on aptamers resulted in nonspecific cell anchoring in control cell lines, affecting the specificity of aptamers. Overall, we found that using a chemical modification strategy could enhance the deep tumor penetration ability of aptamers and validate their effectiveness in vivo. This approach has significant practical applications in targeted drug delivery for cancer treatment.
Collapse
Affiliation(s)
- Yingying Li
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Chi Zhang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ting Fu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Xue-Qiang Wang
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Weihong Tan
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
30
|
Gamboa J, Lourenço P, Cruz C, Gallardo E. Aptamers for the Delivery of Plant-Based Compounds: A Review. Pharmaceutics 2024; 16:541. [PMID: 38675202 PMCID: PMC11053555 DOI: 10.3390/pharmaceutics16040541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Natural compounds have a high potential for the treatment of various conditions, including infections, inflammatory diseases, and cancer. However, they usually present poor pharmacokinetics, low specificity, and even toxicity, which limits their use. Therefore, targeted drug delivery systems, typically composed of a carrier and a targeting ligand, can enhance natural product selectivity and effectiveness. Notably, aptamers-short RNA or single-stranded DNA molecules-have gained attention as promising ligands in targeted drug delivery since they are simple to synthesize and modify, and they present high tissue permeability, stability, and a wide array of available targets. The combination of natural products, namely plant-based compounds, with a drug delivery system utilizing aptamers as targeting agents represents an emerging strategy that has the potential to broaden its applications. This review discusses the potential of aptamers as targeting agents in the delivery of natural compounds, as well as new trends and developments in their utilization in the field of medicine.
Collapse
Affiliation(s)
- Joana Gamboa
- Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior (CICS-UBI), Av. Infante D. Henrique, 6201-506 Covilhã, Portugal; (J.G.); (P.L.)
| | - Pedro Lourenço
- Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior (CICS-UBI), Av. Infante D. Henrique, 6201-506 Covilhã, Portugal; (J.G.); (P.L.)
| | - Carla Cruz
- Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior (CICS-UBI), Av. Infante D. Henrique, 6201-506 Covilhã, Portugal; (J.G.); (P.L.)
- Departamento de Química, Faculdade de Ciências, Universidade da Beira Interior, Rua Marquês de Ávila e Bolama, 6201-001 Covilhã, Portugal
| | - Eugenia Gallardo
- Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior (CICS-UBI), Av. Infante D. Henrique, 6201-506 Covilhã, Portugal; (J.G.); (P.L.)
- Laboratório de Fármaco-Toxicologia, UBIMedical, Universidade da Beira Interior, EM506, 6200-000 Covilhã, Portugal
| |
Collapse
|
31
|
Waldron OP, El-Mallah JC, Lochan D, Wen C, Landmesser ME, Asgardoon M, Dawes J, Horchler SN, Schlidt K, Agrawal S, Wang Y, Ravnic DJ. Ushering in the era of regenerative surgery. Minerva Surg 2024; 79:166-182. [PMID: 38088753 DOI: 10.23736/s2724-5691.23.10113-4] [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: 04/10/2024]
Abstract
Tissue loss, irrespective of etiology, often requires extensive reconstruction. In many instances, the need exceeds what current treatments and technologies modern medicine can offer. Tissue engineering has made immense strides within the past few decades due to advances in biologics, biomaterials, and manufacturing. The convergence of these three domains has created limitless potential for future surgical care. Unfortunately, there still exists a disconnect on how to best implant these 'replacement parts' and care for the patient. It is therefore vital to develop paradigms for the integration of advanced surgical and tissue engineering technologies. This paper explores the convergence between tissue engineering and reconstructive surgery. We will describe the clinical problem of tissue loss, discuss currently available solutions, address limitations, and propose processes for integrating surgery and tissue engineering, thereby ushering in the era of regenerative surgery.
Collapse
Affiliation(s)
- Olivia P Waldron
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
| | - Jessica C El-Mallah
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Dev Lochan
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
| | - Connie Wen
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Mary E Landmesser
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Mohammadhossein Asgardoon
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Jazzmyn Dawes
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
| | - Summer N Horchler
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
| | - Kevin Schlidt
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
| | - Shailaja Agrawal
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Yong Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Dino J Ravnic
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA -
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
32
|
Wang SQ, Wang Y, Yang X, Liu Y, Li H, Yang Z, Sun WY, Sessler JL. High-nuclearity Luminescent Lanthanide Nanocages for Tumor Drug Delivery. Angew Chem Int Ed Engl 2024; 63:e202317775. [PMID: 38286749 DOI: 10.1002/anie.202317775] [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: 11/21/2023] [Revised: 12/31/2023] [Accepted: 01/29/2024] [Indexed: 01/31/2024]
Abstract
There is an unmet need for easy-to-visualize drug carriers that can deliver therapeutic cargoes deep into solid tumors. Herein, we report the preparation of ultrasmall luminescent imine-based lanthanide nanocages, Eu60 and Tb60 (collectively Ln60 ), designed to encapsulate anticancer chemotherapeutics for tumor therapy. The as-prepared nanocages possess large cavities suitable for the encapsulation of doxorubicin (DOX), yielding DOX@Ln60 nanocages with diameters around 5 nm. DOX@Ln60 are efficiently internalized by breast cancer cells, allowing the cells to be visualized via the intrinsic luminescent property of Ln(III). Once internalized, the acidic intracellular microenvironment promotes imine bond cleavage and the release of the loaded DOX. DOX@Ln60 inhibits DNA replication and triggers tumor cell apoptosis. In a murine triple negative breast cancer (TNBC) model, DOX@Ln60 was found to inhibit tumor growth with negligible side effects on normal tissues. It proved more effective than various controls, including DOX and Ln60 . The present nanocages thus point the way to the development of precise nanomedicines for tumor imaging and therapy.
Collapse
Affiliation(s)
- Shi-Qing Wang
- College of Chemistry and Materials Engineering, Zhejiang Key Laboratory of Carbon Materials, Wenzhou University, Wenzhou, 325035, China
| | - Yili Wang
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325035, China
| | - Xiaoping Yang
- College of Chemistry and Materials Engineering, Zhejiang Key Laboratory of Carbon Materials, Wenzhou University, Wenzhou, 325035, China
| | - Yong Liu
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325035, China
| | - Huaqiong Li
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325035, China
| | - Zhi Yang
- College of Chemistry and Materials Engineering, Zhejiang Key Laboratory of Carbon Materials, Wenzhou University, Wenzhou, 325035, China
| | - Wei-Yin Sun
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing National Laboratory of Microstructures, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210023, China
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin, 105 E. 24th Street-A5300, Austin, Texas, 78712-1224, United States
| |
Collapse
|
33
|
Shi Y, Zhen X, Zhang Y, Li Y, Koo S, Saiding Q, Kong N, Liu G, Chen W, Tao W. Chemically Modified Platforms for Better RNA Therapeutics. Chem Rev 2024; 124:929-1033. [PMID: 38284616 DOI: 10.1021/acs.chemrev.3c00611] [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: 01/30/2024]
Abstract
RNA-based therapies have catalyzed a revolutionary transformation in the biomedical landscape, offering unprecedented potential in disease prevention and treatment. However, despite their remarkable achievements, these therapies encounter substantial challenges including low stability, susceptibility to degradation by nucleases, and a prominent negative charge, thereby hindering further development. Chemically modified platforms have emerged as a strategic innovation, focusing on precise alterations either on the RNA moieties or their associated delivery vectors. This comprehensive review delves into these platforms, underscoring their significance in augmenting the performance and translational prospects of RNA-based therapeutics. It encompasses an in-depth analysis of various chemically modified delivery platforms that have been instrumental in propelling RNA therapeutics toward clinical utility. Moreover, the review scrutinizes the rationale behind diverse chemical modification techniques aiming at optimizing the therapeutic efficacy of RNA molecules, thereby facilitating robust disease management. Recent empirical studies corroborating the efficacy enhancement of RNA therapeutics through chemical modifications are highlighted. Conclusively, we offer profound insights into the transformative impact of chemical modifications on RNA drugs and delineates prospective trajectories for their future development and clinical integration.
Collapse
Affiliation(s)
- Yesi Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xueyan Zhen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yiming Zhang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 310058, China
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
34
|
Zhang C, Tang S, Wang M, Li L, Li J, Wang D, Mi X, Zhang Y, Tan X, Yue S. "Triple-Punch" Strategy Exosome-Mimetic Nanovesicles for Triple Negative Breast Cancer Therapy. ACS NANO 2024. [PMID: 38335265 DOI: 10.1021/acsnano.3c10568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most malignant breast cancer, with high rates of relapse and metastasis. Because of the nonspecific targeting of chemotherapy and insurmountable aggressiveness, TNBC therapy lacks an effective strategy. Exosomes have been reported as an efficient drug delivery system (DDS). CD82 is a tumor metastasis inhibitory molecule that is enriched in exosomes. Aptamer AS1411 specifically targets TNBC cells due to its high expression of nucleolin. We generated a "triple-punch" cell membrane-derived exosome-mimetic nanovesicle system that integrated with CD82 overexpression, AS1411 conjugation, and doxorubicin (DOX) delivery. CD82 enrichment effectively inhibits the migration of TNBC cells. AS1411 conjugation specifically targets TNBC cells. DOX loading effectively inhibits proliferation and induces apoptosis of TNBC cells. Our results demonstrate a system of exosome-mimetic nanovesicles with "triple-punch" that may facilitate TNBC therapeutics.
Collapse
Affiliation(s)
- Chenhong Zhang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shuangshuang Tang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Meilin Wang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Luhan Li
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Jun Li
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Dekun Wang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xue Mi
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Yuying Zhang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Xiaoyue Tan
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shijing Yue
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
35
|
Feng K, Li X, Bai Y, Zhang D, Tian L. Mechanisms of cancer cell death induction by triptolide: A comprehensive overview. Heliyon 2024; 10:e24335. [PMID: 38293343 PMCID: PMC10826740 DOI: 10.1016/j.heliyon.2024.e24335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
The need for naturally occurring constituents is driven by the rise in the cancer prevalence and the unpleasant side effects associated with chemotherapeutics. Triptolide, the primary active component of "Tripterygium Wilfordii", has exploited for biological mechanisms and therapeutic potential against various tumors. Based on the recent pre-clinical investigations, triptolide is linked to the induction of death of cancerous cells by triggering cellular apoptosis via inhibiting heat shock protein expression (HSP70), and cyclin dependent kinase (CDKs) by up regulating expression of P21. MKP1, histone methyl transferases and RNA polymerases have all recently identified as potential targets of triptolide in cells. Autophagy, AKT signaling pathway and various pathways involving targeted proteins such as A-disintegrin & metalloprotease-10 (ADAM10), Polycystin-2 (PC-2), dCTP pyro-phosphatase 1 (DCTP1), peroxiredoxin-I (Prx-I), TAK1 binding protein (TAB1), kinase subunit (DNA-PKcs) and the xeroderma-pigmentosum B (XPB or ERCC3) have been exploited. Besides that, triptolide is responsible for enhancing the effectiveness of various chemotherapeutics. In addition, several triptolide moieties, including minnelide and LLDT8, have progressed in investigations on humans for the treatment of cancer. Targeted strategies, such as triptolide conjugation with ligands or triptolide loaded nano-carriers, are efficient techniques to confront toxicities associated with triptolide. We expect and anticipate that advances in near future, regarding combination therapies of triptolide, might be beneficial against cancerous cells.
Collapse
Affiliation(s)
- Ke Feng
- Department of General Surgery, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| | - Xiaojiang Li
- Department of General Surgery, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| | - Yuzhuo Bai
- Department of Breast and Thyroid Surgery Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| | - Dawei Zhang
- Department of General Surgery Baishan Hospital of Traditional Chinese Medicine, Baishan, 134300, China
| | - Lin Tian
- Department of Lung Oncology, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130000, China
| |
Collapse
|
36
|
Liu R, Liu Z, Chen M, Xing H, Zhang P, Zhang J. Cooperatively designed aptamer-PROTACs for spatioselective degradation of nucleocytoplasmic shuttling protein for enhanced combinational therapy. Chem Sci 2023; 15:134-145. [PMID: 38131089 PMCID: PMC10732009 DOI: 10.1039/d3sc04249a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Nucleocytoplasmic shuttling proteins (NSPs) have emerged as a promising class of therapeutic targets for many diseases. However, most NSPs-based therapies largely rely on small-molecule inhibitors with limited efficacy and off-target effects. Inspired by proteolysis targeting chimera (PROTAC) technology, we report a new archetype of PROTAC (PS-ApTCs) by introducing a phosphorothioate-modified aptamer to a CRBN ligand, realizing tumor-targeting and spatioselective degradation of NSPs with improved efficacy. Using nucleolin as a model, we demonstrate that PS-ApTCs is capable of effectively degrading nucleolin in the target cell membrane and cytoplasm but not in the nucleus, through the disruption of nucleocytoplasmic shuttling. Moreover, PS-ApTCs exhibits superior antiproliferation, pro-apoptotic, and cell cycle arrest potencies. Importantly, we demonstrate that a combination of PS-ApTCs-mediated nucleolin degradation with aptamer-drug conjugate-based chemotherapy enables a synergistic effect on tumor inhibition. Collectively, PS-ApTCs could expand the PROTAC toolbox to more targets in subcellular localization and accelerate the discovery of new combinational therapeutic approaches.
Collapse
Affiliation(s)
- Ran Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Zheng Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Mohan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| | - Hang Xing
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, College of Chemistry and Chemical Engineering, Hunan University Changsha 410082 China
| | - Penghui Zhang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou 310022 China
| | - Jingjing Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University Nanjing 210023 China
| |
Collapse
|
37
|
Liu J, Ren Z, Sun Y, Xu L, Wei D, Tan W, Ding D. Investigation of the Relationship between Aptamers' Targeting Functions and Human Plasma Proteins. ACS NANO 2023; 17:24329-24342. [PMID: 38044589 DOI: 10.1021/acsnano.3c10238] [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: 12/05/2023]
Abstract
Aptamers are single-stranded DNA or RNA molecules capable of recognizing targets via specific three-dimensional structures. Taking advantage of this unique targeting function, aptamers have been extensively applied to bioanalysis and disease theranostics. However, the targeting functionality of aptamers in the physiological milieu is greatly impeded compared with their in vitro applications. To investigate the physiological factors that adversely affect the in vivo targeting ability of aptamers, we herein systematically studied the interactions between human plasma proteins and aptamers and the specific effects of plasma proteins on aptamer targeting. Microscale thermophoresis and flow cytometry analysis showed that plasma interacted with aptamers, restricting their affinity toward targeted tumor cells. Further pull-down assay and proteomic identification verified that the interactions between aptamers and plasma proteins were mainly involved in complement activation and immune response as well as showed structure-selective and sequence-specific features. Particularly, the fibronectin 1 (FN1) protein showed dramatically specific interactions with nucleolin (NCL) targeting aptamer AS1411. The competitive binding between FN1 and NCL almost deprived the AS1411 aptamer's targeting ability in vivo. In order to maintain the targeting function in the physiological milieu, a series of optimizations were performed via the chemical modifications of AS1411 aptamer, and 3'-terminal pegylation was demonstrated to be resistant to the interaction with FN1, leading to improved tumor-targeting effects. This work emphasizes the physiological environment influences on aptamers targeting functionality and suggests that rational design and modification of aptamers to minimize the nonspecific interaction with plasma proteins might be effective to maintain aptamer functionality in future clinical uses.
Collapse
Affiliation(s)
- Jia Liu
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Zhiqiang Ren
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai 200234, People's Republic of China
| | - Yang Sun
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Liujun Xu
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Dali Wei
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, People's Republic of China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Ding Ding
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| |
Collapse
|
38
|
Liu X, Sun M, Pu F, Ren J, Qu X. Transforming Intratumor Bacteria into Immunopotentiators to Reverse Cold Tumors for Enhanced Immuno-chemodynamic Therapy of Triple-Negative Breast Cancer. J Am Chem Soc 2023; 145:26296-26307. [PMID: 37987621 DOI: 10.1021/jacs.3c09472] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Immunotherapy of triple-negative breast cancer (TNBC) has an unsatisfactory therapeutic outcome due to an immunologically "cold" microenvironment. Fusobacterium nucleatum (F. nucleatum) was found to be colonized in triple-negative breast tumors and was responsible for the immunosuppressive tumor microenvironment and tumor metastasis. Herein, we constructed a bacteria-derived outer membrane vesicle (OMV)-coated nanoplatform that precisely targeted tumor tissues for dual killing of F. nucleatum and cancer cells, thus transforming intratumor bacteria into immunopotentiators in immunotherapy of TNBC. The as-prepared nanoparticles efficiently induced immunogenic cell death through a Fenton-like reaction, resulting in enhanced immunogenicity. Meanwhile, intratumoral F. nucleatum was killed by metronidazole, resulting in the release of pathogen-associated molecular patterns (PAMPs). PAMPs cooperated with OMVs further facilitated the maturation of dendritic cells and subsequent T-cell infiltration. As a result, the "kill two birds with one stone" strategy warmed up the cold tumor environment, maximized the antitumor immune response, and achieved efficient therapy of TNBC as well as metastasis prevention. Overall, this strategy based on a microecology distinction in tumor and normal tissue as well as microbiome-induced reversal of cold tumors provides new insight into the precise and efficient immune therapy of TNBC.
Collapse
Affiliation(s)
- Xuemeng Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Jilin, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Anhui, Hefei 230026, P.R. China
| | - Mengyu Sun
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Jilin, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Anhui, Hefei 230026, P.R. China
| | - Fang Pu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Jilin, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Anhui, Hefei 230026, P.R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Jilin, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Anhui, Hefei 230026, P.R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Jilin, Changchun 130022, P.R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Anhui, Hefei 230026, P.R. China
| |
Collapse
|
39
|
Tian Q, Zhang P, Wang Y, Si Y, Yin D, Weber CR, Fishel ML, Pollok KE, Qiu B, Xiao F, Chong AS. A novel triptolide analog downregulates NF-κB and induces mitochondrial apoptosis pathways in human pancreatic cancer. eLife 2023; 12:e85862. [PMID: 37877568 PMCID: PMC10861173 DOI: 10.7554/elife.85862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 10/24/2023] [Indexed: 10/26/2023] Open
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related death worldwide, and despite advancements in disease management, the 5 -year survival rate stands at only 12%. Triptolides have potent anti-tumor activity against different types of cancers, including pancreatic cancer, however poor solubility and toxicity limit their translation into clinical use. We synthesized a novel pro-drug of triptolide, (E)-19-[(1'-benzoyloxy-1'-phenyl)-methylidene]-Triptolide (CK21), which was formulated into an emulsion for in vitro and in vivo testing in rats and mice, and used human pancreatic cancer cell lines and patient-derived pancreatic tumor organoids. A time-course transcriptomic profiling of tumor organoids treated with CK21 in vitro was conducted to define its mechanism of action, as well as transcriptomic profiling at a single time point post-CK21 administration in vivo. Intravenous administration of emulsified CK21 resulted in the stable release of triptolide, and potent anti-proliferative effects on human pancreatic cancer cell lines and patient-derived pancreatic tumor organoids in vitro, and with minimal toxicity in vivo. Time course transcriptomic profiling of tumor organoids treated with CK21 in vitro revealed <10 differentially expressed genes (DEGs) at 3 hr and ~8,000 DEGs at 12 hr. Overall inhibition of general RNA transcription was observed, and Ingenuity pathway analysis together with functional cellular assays confirmed inhibition of the NF-κB pathway, increased oxidative phosphorylation and mitochondrial dysfunction, leading ultimately to increased reactive oxygen species (ROS) production, reduced B-cell-lymphoma protein 2 (BCL2) expression, and mitochondrial-mediated tumor cell apoptosis. Thus, CK21 is a novel pro-drug of triptolide that exerts potent anti-proliferative effects on human pancreatic tumors by inhibiting the NF-κB pathway, leading ultimately to mitochondrial-mediated tumor cell apoptosis.
Collapse
Affiliation(s)
- Qiaomu Tian
- Department of Surgery, The University of ChicagoChicagoUnited States
| | - Peng Zhang
- Cinkate Pharmaceutical Corp, ZhangJiang DistrictShanghaiChina
| | - Yihan Wang
- Department of Surgery, The University of ChicagoChicagoUnited States
| | - Youhui Si
- Department of Surgery, The University of ChicagoChicagoUnited States
| | - Dengping Yin
- Department of Surgery, The University of ChicagoChicagoUnited States
| | | | - Melissa L Fishel
- Department of Pediatrics, Indiana UniversityIndianapolisUnited States
| | - Karen E Pollok
- Department of Pediatrics, Indiana UniversityIndianapolisUnited States
| | - Bo Qiu
- Cinkate Pharmaceutical Corp, ZhangJiang DistrictShanghaiChina
| | - Fei Xiao
- Cinkate Pharmaceutical Corp, ZhangJiang DistrictShanghaiChina
| | - Anita S Chong
- Department of Surgery, The University of ChicagoChicagoUnited States
| |
Collapse
|
40
|
Yang LT, Abudureheman T, Zheng WW, Zhou H, Chen J, Duan CW, Chen KM. A novel His-tag-binding aptamer for recombinant protein detection and T cell-based immunotherapy. Talanta 2023; 263:124722. [PMID: 37247456 DOI: 10.1016/j.talanta.2023.124722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/04/2023] [Accepted: 05/22/2023] [Indexed: 05/31/2023]
Abstract
Screening novel aptamers for recombinant protein detection is of great significance in industrial mass production of antibody drugs. In addition, construction of structurally stable bispecific circular aptamers (bc-apts) may provide a tumor-targeted treatment strategy by simultaneously binding two different cell types. In this study, we obtained a high-affinity hexahistidine tag (His-tag)-binding aptamer 20S and explored its application in recombinant protein detection and T cell-based immunotherapy. We developed a new molecular beacon (MB) 20S-MB to detect His-tagged proteins in vitro and in vivo with high sensitivity and specificity, and the results showed high consistency with the enzyme-linked immunosorbent assay (ELISA). Moreover, we constructed two kinds of bc-apts by cyclizing 20S or another His-tag-binding aptamer, 6H5-MU, with Sgc8, which specifically recognizes protein tyrosine kinase 7 (PTK7) on tumor cells. After forming a complex with His-tagged OKT3, an anti-CD3 antibody for T cell activation, we utilized these aptamer-antibody complexes (ap-ab complex) to enhance cytotoxicity of T cells by linking T cells and target cells together, and 20S-sgc8 exhibited antitumor efficacy superior to that of 6H5-sgc8. In conclusion, we screened a novel His-tag-binding aptamer and used it to construct a new type of MB for rapid detection of recombinant proteins, as well as establish a feasible approach for T cell-based immunotherapy.
Collapse
Affiliation(s)
- Li-Ting Yang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tuersunayi Abudureheman
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Wei Zheng
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hang Zhou
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Chen
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Cai-Wen Duan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China; Fujian Branch of Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, And Fujian Children's Hospital, China; Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate, National Health Commission, Fujian Maternity and Child Health Hospital, China.
| | - Kai-Ming Chen
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Fujian Branch of Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, And Fujian Children's Hospital, China.
| |
Collapse
|
41
|
Chen S, Cao R, Xiang L, Li Z, Chen H, Zhang J, Feng X. Research progress in nucleus-targeted tumor therapy. Biomater Sci 2023; 11:6436-6456. [PMID: 37609783 DOI: 10.1039/d3bm01116j] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
The nucleus is considered the most important organelle in the cell as it plays a central role in controlling cell reproduction, metabolism, and the cell cycle. The successful delivery of drugs into the nucleus can achieve excellent therapeutic effects, which reveals the potential of nucleus-targeted therapy in precision medicine. However, the transportation of therapeutics into the nucleus remains a significant challenge due to various biological barriers. Herein, we summarize the recent progress in the nucleus-targeted drug delivery system (NDDS). The structures of the nucleus and nuclear envelope are first described in order to understand the mechanisms by which drugs cross the nuclear envelope. Then, various drug delivery strategies based on the mechanisms and their applications are discussed. Finally, the challenges and solutions in the field of nucleus-targeted drug delivery are raised for developing a more efficient NDDS and promoting its clinical transformation.
Collapse
Affiliation(s)
- Shaofeng Chen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Rumeng Cao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Ling Xiang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Ziyi Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Hui Chen
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Jiumeng Zhang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| | - Xuli Feng
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, P. R. China.
| |
Collapse
|
42
|
Xiao CD, Zhong MQ, Gao Y, Yang ZL, Jia MH, Hu XH, Xu Y, Shen XC. A Unique G-Quadruplex Aptamer: A Novel Approach for Cancer Cell Recognition, Cell Membrane Visualization, and RSV Infection Detection. Int J Mol Sci 2023; 24:14344. [PMID: 37762645 PMCID: PMC10531985 DOI: 10.3390/ijms241814344] [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: 08/23/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Surface staining has emerged as a rapid technique for applying external stains to trace cellular identities in diverse populations. In this study, we developed a distinctive aptamer with selective binding to cell surface nucleolin (NCL), bypassing cytoplasmic internalization. Conjugation of the aptamer with a FAM group facilitated NCL visualization on live cell surfaces with laser confocal microscopy. To validate the aptamer-NCL interaction, we employed various methods, including the surface plasmon resonance, IHC-based flow cytometry, and electrophoretic mobility shift assay. The G-quadruplex formations created by aptamers were confirmed with a nuclear magnetic resonance and an electrophoretic mobility shift assay utilizing BG4, a G-quadruplex-specific antibody. Furthermore, the aptamer exhibited discriminatory potential in distinguishing between cancerous and normal cells using flow cytometry. Notably, it functioned as a dynamic probe, allowing real-time monitoring of heightened NCL expression triggered by a respiratory syncytial virus (RSV) on normal cell surfaces. This effect was subsequently counteracted with dsRNA transfection and suppressed the NCL expression; thus, emphasizing the dynamic attributes of the probe. These collective findings highlight the robust versatility of our aptamer as a powerful tool for imaging cell surfaces, holding promising implications for cancer cell identification and the detection of RSV infections.
Collapse
Affiliation(s)
- Chao-Da Xiao
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (M.-Q.Z.); (Y.G.); (Z.-L.Y.); (M.-H.J.); (X.-H.H.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Ming-Qing Zhong
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (M.-Q.Z.); (Y.G.); (Z.-L.Y.); (M.-H.J.); (X.-H.H.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Yue Gao
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (M.-Q.Z.); (Y.G.); (Z.-L.Y.); (M.-H.J.); (X.-H.H.)
| | - Zheng-Lin Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (M.-Q.Z.); (Y.G.); (Z.-L.Y.); (M.-H.J.); (X.-H.H.)
| | - Meng-Hao Jia
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (M.-Q.Z.); (Y.G.); (Z.-L.Y.); (M.-H.J.); (X.-H.H.)
| | - Xiao-Hui Hu
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (M.-Q.Z.); (Y.G.); (Z.-L.Y.); (M.-H.J.); (X.-H.H.)
| | - Yan Xu
- Division of Chemistry, Department of Medical Sciences, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan;
| | - Xiang-Chun Shen
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; (M.-Q.Z.); (Y.G.); (Z.-L.Y.); (M.-H.J.); (X.-H.H.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| |
Collapse
|
43
|
Chang R, Li T, Fu Y, Chen Z, He Y, Sun X, Deng Y, Zhong Y, Xie Z, Yang Y, Liu J, Chen X, Liu H, Zhao Y. A PD-L1 targeting nanotheranostic for effective photoacoustic imaging guided photothermal-immunotherapy of tumor. J Mater Chem B 2023; 11:8492-8505. [PMID: 37594411 DOI: 10.1039/d3tb00221g] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Tumor immunotherapy has been partly effective for specific cancers. However, problems such as low immune response, limited antitumor effectiveness, and high antibody costs still persist. Synergistic therapeutic approaches, such as immune checkpoint inhibition in conjunction with photothermal therapy and photoacoustic imaging, are expected to provide approaches for more precise and efficient immunotherapy of tumors. Furthermore, developing alternatives for antibodies, such as PD-L1 aptamers and nanocarriers, would reduce the cost of tumor immunotherapy. Herein, we develop a PD-L1-targeting nanotheranostic to block immune checkpoints for synergistic photothermal-immunotherapy against tumors, along with effective photoacoustic (PA) imaging. The nanotheranostic is synthesized by the modification of gold nanorods (GNRs) with the PD-L1 aptamer (APDL1), which can sensitively and specifically recognize PD-L1 on the tumor cell surface, and mediate nanoparticle accumulation and strong PA signals in tumors. The aptamer is released from GNR through a competition of glutathione (GSH) and is then functionalized as a PD-L1 blockade. In collaboration with the concurrent photothermal therapy, antitumor immunity is significantly augmented by enhancing the filtration of matured dendritic cells and suppressing regulatory T cells, followed by the activation of cytotoxic T cells and inhibition of T cell exhaustion. Such a nanotheranostic modality effectively suppresses tumor growth in mice, representing an appealing platform for both biological imaging and photoimmunotherapy of tumors.
Collapse
Affiliation(s)
- Ruimin Chang
- Department of Dermatology, Xiangya Clinical Research Center for Cancer Immunotherapy, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Tan Li
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410078, China.
| | - Yao Fu
- Department of Dermatology, Xiangya Clinical Research Center for Cancer Immunotherapy, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zeyu Chen
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Yilang He
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410078, China.
| | - Xin Sun
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Yiyi Deng
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410078, China.
| | - Yanqing Zhong
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410078, China.
| | - Zuozhong Xie
- Department of Dermatology, Xiangya Clinical Research Center for Cancer Immunotherapy, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Yang Yang
- College of Biotechnology, Tianjin University of Science & Technology, No. 29, 13th Avenue, TEDA, Tianjin, 300457, China
| | - Jing Liu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410078, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Clinical Research Center for Cancer Immunotherapy, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Hong Liu
- Department of Dermatology, Xiangya Clinical Research Center for Cancer Immunotherapy, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Yuetao Zhao
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
44
|
Pleiko K, Haugas M, Parfejevs V, Pantelejevs T, Parisini E, Teesalu T, Riekstina U. Targeting triple-negative breast cancer cells with a β1-integrin binding aptamer. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:871-884. [PMID: 37680989 PMCID: PMC10481362 DOI: 10.1016/j.omtn.2023.08.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/14/2023] [Indexed: 09/09/2023]
Abstract
Targeted therapies have increased the treatment options for triple-negative breast cancer patients. However, the paucity of targetable biomarkers and tumor heterogeneity have limited the ability of precision-guided interventions to live up to their full potential. As affinity-targeting ligands, aptamers show high selectivity toward target molecules. Compared with antibodies, aptamers have lower molecular weight, increased stability during transportation, reduced immunogenicity, and increased tissue uptake. Recently, we reported discovery of the GreenB1 aptamer, which is internalized in cultured triple-negative MDA-MB-231 human breast cancer cells. We show that the GreenB1 aptamer specifically targets β1-integrin, a protein linked previously to breast cancer cell invasiveness and migration. Aptamer binds to β1-integrin with low nanomolar affinity. Our findings suggest potential applications for GreenB1-guided precision agents for diagnosis and therapy of cancers overexpressing β1-integrin.
Collapse
Affiliation(s)
- Karlis Pleiko
- Faculty of Medicine, University of Latvia, House of Science, Jelgavas Str. 3, 1004 Riga, Latvia
- Laboratory of Precision- and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Maarja Haugas
- Laboratory of Precision- and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Vadims Parfejevs
- Faculty of Medicine, University of Latvia, House of Science, Jelgavas Str. 3, 1004 Riga, Latvia
| | | | - Emilio Parisini
- Latvian Institute of Organic Synthesis, Aizkraukles 21, 1006 Riga, Latvia
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Tambet Teesalu
- Laboratory of Precision- and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
- Materials Research Laboratory, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Una Riekstina
- Faculty of Medicine, University of Latvia, House of Science, Jelgavas Str. 3, 1004 Riga, Latvia
| |
Collapse
|
45
|
Cong Y, Zhang SY, Tang PYZ, Li HM, Liu X, Zhao W, Tang YJ. Conjugating 4β-NH-(5-Aminoindazole)-podophyllotoxin and Galectin-1-Targeted Aptamer for Synergistic Chemo-Immunotherapy of Hepatocellular Carcinoma. Adv Healthc Mater 2023; 12:e2203144. [PMID: 37141264 DOI: 10.1002/adhm.202203144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/28/2023] [Indexed: 05/05/2023]
Abstract
By conjugating a chemotherapeutic candidate drug 4β-NH-(5-aminoindazole)-podophyllotoxin (βIZP) and an immunosuppressive protein galectin-1 targeted aptamer AP74, a chemo-immunotherapy molecule (AP74-βIZP) is developed against liver cancer. AP74-βIZP can target galectin-1 and enrich the tumor microenvironment to improve the tumor inhibition ratio by 6.3%, higher than that of βIZP in a HepG2 xenograft model. In safety evaluation, βIZP cannot be released from AP74-βIZP in normal tissues with low glutathione level. Therefore, the degrees of organs injury and myelosuppression after the treatment with AP74-βIZP are lower than those with βIZP. After 21 d treatment at a drug dose of 5 mg kg-1 , AP74-βIZP does not cause weight loss in mice, while the weight is significantly reduced by 24% and 14% from oxaliplatin and βIZP, respectively. In immune synergy, AP74-IZP enhances CD4/CD8 cell infiltration to promote the expression of cell factor (i.e., IL-2, TNF-α, and IFN-γ), which further improves the antitumor activity. The tumor inhibition ratio of AP74-βIZP is 70.2%, which is higher than that of AP74 (35.2%) and βIZP (48.8%). Because of the dual effects of chemotherapy and immunotherapy, AP74-βIZP exhibits superior activity and lower toxicity. The approach developed in this work could be applicable to other chemotherapy drugs.
Collapse
Affiliation(s)
- Ying Cong
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| | - Shu-Yue Zhang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, China
| | | | - Hong-Mei Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| | - Xue Liu
- Jinan Food and Drug Inspection and Testing Center, Jinan, 250101, China
| | - Wei Zhao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| |
Collapse
|
46
|
Zhu S, Zhang T, Gao H, Jin G, Yang J, He X, Guo H, Xu F. Combination Therapy of Lox Inhibitor and Stimuli-Responsive Drug for Mechanochemically Synergistic Breast Cancer Treatment. Adv Healthc Mater 2023; 12:e2300103. [PMID: 37099721 DOI: 10.1002/adhm.202300103] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 04/21/2023] [Indexed: 04/28/2023]
Abstract
Chemotherapy based on small molecule drugs, hormones, cycline kinase inhibitors, and monoclonal antibodies has been widely used for breast cancer treatment in the clinic but with limited efficacy, due to the poor specificity and tumor microenvironment (TME)-caused diffusion barrier. Although monotherapies targeting biochemical cues or physical cues in the TME have been developed, none of them can cope with the complex TME, while mechanochemical combination therapy remains largely to be explored. Herein, a combination therapy strategy based on an extracellular matrix (ECM) modulator and TME-responsive drug for the first attempt of mechanochemically synergistic treatment of breast cancer is developed. Specifically, based on overexpressed NAD(P)H quinone oxidoreductase 1 (NQO1) in breast cancer, a TME-responsive drug (NQO1-SN38) is designed and it is combined with the inhibitor (i.e., β-Aminopropionitrile, BAPN) for Lysyl oxidases (Lox) that contributes to the tumor stiffness, for mechanochemical therapy. It is demonstrated that NQO1 can trigger the degradation of NQO1-SN38 and release SN38, showing nearly twice tumor inhibition efficiency compared with SN38 treatment in vitro. Lox inhibition with BAPN significantly reduces collagen deposition and enhances drug penetration in tumor heterospheroids in vitro. It is further demonstrated that the mechanochemical therapy showed outstanding therapeutic efficacy in vivo, providing a promising approach for breast cancer therapy.
Collapse
Affiliation(s)
- Shanshan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Tian Zhang
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Huan Gao
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Guorui Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Jin Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Xiaocong He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Hui Guo
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
47
|
Ai L, Jiang X, Zhang K, Cui C, Liu B, Tan W. Tools and techniques for the discovery of therapeutic aptamers: recent advances. Expert Opin Drug Discov 2023; 18:1393-1411. [PMID: 37840268 DOI: 10.1080/17460441.2023.2264187] [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: 03/15/2023] [Accepted: 09/25/2023] [Indexed: 10/17/2023]
Abstract
INTRODUCTION The pursuit of novel therapeutic agents for serious diseases such as cancer has been a global endeavor. Aptamers characteristic of high affinity, programmability, low immunogenicity, and rapid permeability hold great promise for the treatment of diseases. Yet obtaining the approval for therapeutic aptamers remains challenging. Consequently, researchers are increasingly devoted to exploring innovative strategies and technologies to advance the development of these therapeutic aptamers. AREAS COVERED The authors provide a comprehensive summary of the recent progress of the SELEX (Systematic Evolution of Ligands by EXponential enrichment) technique, and how the integration of modern tools has facilitated the identification of therapeutic aptamers. Additionally, the engineering of aptamers to enhance their functional attributes, such as inhibiting and targeting, is discussed, demonstrating the potential to broaden their scope of utility. EXPERT OPINION The grand potential of aptamers and the insufficient development of relevant drugs have spurred countless efforts for stimulating their discovery and application in the therapeutic field. While SELEX techniques have undergone significant developments with the aid of advanced analysis instruments and ingeniously updated aptameric engineering strategies, several challenges still impede their clinical translation. A key challenge lies in the insufficient understanding of binding conformation and susceptibility to degradation under physiological conditions. Despite the hurdles, our opinion is optimistic. With continued progress in overcoming these obstacles, the widespread utilization of aptamers for clinical therapy is envisioned to become a reality soon.
Collapse
Affiliation(s)
- Lili Ai
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, The People's Republic of China
| | - Xinyi Jiang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, The People's Republic of China
| | - Kejing Zhang
- Department of Geriatrics and Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, The People's Republic of China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, The People's Republic of China
| | - Cheng Cui
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, The People's Republic of China
| | - Bo Liu
- Department of Geriatrics and Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, The People's Republic of China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, The People's Republic of China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, The People's Republic of China
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, The People's Republic of China
| |
Collapse
|
48
|
Cai J, Zhong M, Xu J, Cheng H, Xu S. Codelivery of triptolide and IFN-γ to boost antitumor immunity for triple-negative breast cancer. Int Immunopharmacol 2023; 120:110346. [PMID: 37210915 DOI: 10.1016/j.intimp.2023.110346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 05/23/2023]
Abstract
Triple-negative breast cancer (TNBC) is a specific type of breast cancer that exhibits poor prognosis and complex tumor heterogeneity. The unique immune tumor microenvironment reveals great potential of immunotherapy in TNBC. Triptolide, a potential regulator of immune-related signaling, has shown potent antitumor activity in TNBC. However, the molecular mechanism of triptolide in TNBC is still controversial. This study identified interferon-γ (IFN-γ) as a therapeutical target of triptolide based on the analysis of prognostic biomarkers in TNBC. IFN-γ is an important component of immunotherapy and contributes to antitumor immune activation. Triptolide was found to significantly reverse the IFN-γ-inducible programmed death-ligand 1 (PD-L1) in TNBC. The combined treatment of triptolide and IFN-γ in a hydrogel delivery system remarkably induced the cytotoxic CD8 + T lymphocytes activation, showing a synergistic effect on the potent tumor inhibition.
Collapse
Affiliation(s)
- Jianya Cai
- Department of Surgery, Quanzhou Medical College, Quanzhou, China
| | - Minjie Zhong
- Department of Surgery, The Second Clinical Medical College of Fujian Medical University, Quanzhou, China
| | - Jianhua Xu
- Department of Surgery, The Second Clinical Medical College of Fujian Medical University, Quanzhou, China
| | - Hongwei Cheng
- Center of molecular imaging translational medicine, School of public health, Xiamen University, Xiamen, China
| | - Shuangta Xu
- Department of Surgery, The Second Clinical Medical College of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
49
|
Farahbakhsh Z, Zamani M, Nasirian V, Shariati L, Kermani S, Karizmeh MS, Rafienia M. An insight into fluorescence and magnetic resonance bioimaging using a multifunctional polyethyleneimine-passivated gadocarbon dots nanoconstruct assembled with AS1411. Mikrochim Acta 2023; 190:275. [PMID: 37358641 DOI: 10.1007/s00604-023-05853-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/29/2023] [Indexed: 06/27/2023]
Abstract
A nanoassembly of PEI-passivated Gd@CDs, a type of aptamer, is presented which was designed and characterized in order to target specific cancer cells based on their recognition of the receptor nucleolin (NCL), which is overexpressed on the cell membrane of breast cancer cells for fluorescence and magnetic resonance imaging and treatment. Using hydrothermal methods, Gd-doped nanostructures were synthesized, then modified by a two-step chemical procedure for subsequent applications: the passivating of Gd@CDs with branched polyethyleneimine (PEI) (to form Gd@CDs-PEI1 and Gd@CDs-PEI2), and using AS1411 aptamer (AS) as a DNA-targeted molecule (to generate AS/Gd@CDs-PEI1 and AS/Gd@CDs-PEI2). Consequently, these nanoassemblies were constructed as a result of electrostatic interactions between cationic Gd@CDs-passivated PEI and AS aptamers, offering efficient multimodal targeting nanoassemblies for cancer cell detection. It has been demonstrated through in vitro studies that both types of AS-conjugated nanoassemblies are highly biocompatible, have high cellular uptake efficiency (equivalent concentration of AS: 0.25 μΜ), and enable targeted fluorescence imaging in nucleolin-positive MCF7 and MDA-MB-231 cancer cells compared to MCF10-A normal cells. Importantly, the as-prepared Gd@CDs, Gd@CDs-PEI1, and Gd@CDs-PEI2 exhibit higher longitudinal relaxivity values (r1) compared with the commercial Gd-DTPA, equal to 5.212, 7.488, and 5.667 mM-1s-1, respectively. Accordingly, it is concluded that the prepared nanoassemblies have the potential to become excellent candidates for cancer targeting and fluorescence/MR imaging agents, which can be applied to cancer imaging and personalized nanomedicine.
Collapse
Affiliation(s)
- Zohreh Farahbakhsh
- Department of Biology, Nourdanesh Institute of Higher Education, Meymeh, Isfahan, Iran
- Department of Medical Parasitology and Mycology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Biosensor Research Center (BRC), Isfahan University of Medical Sciences (IUMS), Isfahan, Iran
| | - Mohammadreza Zamani
- Department of Biology, Nourdanesh Institute of Higher Education, Meymeh, Isfahan, Iran
| | - Vahid Nasirian
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA
| | - Laleh Shariati
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, IR, Iran
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeed Kermani
- Department of Bioelectric and Biomedical Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Shie Karizmeh
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Rafienia
- Biosensor Research Center (BRC), Isfahan University of Medical Sciences (IUMS), Isfahan, Iran.
| |
Collapse
|
50
|
Ding D, Zhao H, Wei D, Yang Q, Yang C, Wang R, Chen Y, Li L, An S, Xia Q, Huang G, Liu J, Xiao Z, Tan W. The First-in-Human Whole-Body Dynamic Pharmacokinetics Study of Aptamer. RESEARCH (WASHINGTON, D.C.) 2023; 6:0126. [PMID: 37223462 PMCID: PMC10202413 DOI: 10.34133/research.0126] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/30/2023] [Indexed: 05/25/2023]
Abstract
Serving as targeting ligands, aptamers have shown promise in precision medicine. However, the lack of knowledge of the biosafety and metabolism patterns in the human body largely impeded aptamers' clinical translation. To bridge this gap, here we report the first-in-human pharmacokinetics study of protein tyrosine kinase 7 targeted SGC8 aptamer via in vivo PET tracking of gallium-68 (68Ga) radiolabeled aptamers. The specificity and binding affinity of a radiolabeled aptamer, named 68Ga[Ga]-NOTA-SGC8, were maintained as proven in vitro. Further preclinical biosafety and biodistribution evaluation confirmed that aptamers have no biotoxicity, potential mutation risks, or genotoxicity at high dosage (40 mg/kg). Based on this result, a first-in-human clinical trial was approved and carried out to evaluate the circulation and metabolism profiles, as well as biosafety, of the radiolabeled SGC8 aptamer in the human body. Taking advantage of the cutting-edge total-body PET, the aptamers' distribution pattern in the human body was acquired in a dynamic fashion. This study revealed that radiolabeled aptamers are harmless to normal organs and most of them are accumulated in the kidney and cleared from the bladder via urine, which agrees with preclinical studies. Meanwhile, a physiologically based pharmacokinetic model of aptamer was developed, which could potentially predict therapeutic responses and plan personalized treatment strategies. This research studied the biosafety and dynamic pharmacokinetics of aptamers in the human body for the first time, as well as demonstrated the capability of novel molecular imaging fashion in drug development.
Collapse
Affiliation(s)
- Ding Ding
- Institute of Molecular Medicine (IMM), Renji Hospital, State Key Laboratory of Oncogenes and Related Genes,
Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Haitao Zhao
- Department of Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
| | - Dali Wei
- Institute of Molecular Medicine (IMM), Renji Hospital, State Key Laboratory of Oncogenes and Related Genes,
Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qinglai Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, State Key Laboratory of Oncogenes and Related Genes,
Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Center for Molecular Imaging Probes, Cancer Research Institute,
University of South China, Hengyang, Hunan 421001, China
| | - Cai Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, State Key Laboratory of Oncogenes and Related Genes,
Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital,Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering,
Hunan University, Changsha, Hunan 410082, China
| | - Ruowen Wang
- Institute of Molecular Medicine (IMM), Renji Hospital, State Key Laboratory of Oncogenes and Related Genes,
Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yumei Chen
- Department of Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lianghua Li
- Department of Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shuxian An
- Department of Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qian Xia
- Institute of Molecular Medicine (IMM), Renji Hospital, State Key Laboratory of Oncogenes and Related Genes,
Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging,
Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zeyu Xiao
- Institute of Molecular Medicine (IMM), Renji Hospital, State Key Laboratory of Oncogenes and Related Genes,
Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Pharmacology and Chemical Biology,
Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, State Key Laboratory of Oncogenes and Related Genes,
Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital,Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering,
Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|