1
|
Xie W, Sun W, Li Q, Dang Y, Ma L, Liu Y, Zhang H, Qu F, Tan W. Click-constructed modular signal aptamer chimeras enable receptor-independent degradation of membrane proteins. Proc Natl Acad Sci U S A 2025; 122:e2424500122. [PMID: 40388621 DOI: 10.1073/pnas.2424500122] [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: 11/23/2024] [Accepted: 04/08/2025] [Indexed: 05/21/2025] Open
Abstract
Cell-membrane proteins are critical mediators of signal transduction, playing essential roles in disease occurrence and progression. The emerging LYTACs (Lysosome-targeting chimeras) technology combines drug-targeting strategies with lysosomal degradation, providing a novel approach to drug development and offering new possibilities for disease therapy. However, the clinical applicability of current LYTAC degraders is limited by the variable expression of lysosome-targeting receptors (LTRs) in tissues. To overcome this limitation, we herein hijacked a YXXØ sorting signal that derived from lysosome-associated membrane protein 2a (LAMP-2a) to develop a signal aptamer platform (SApt), which exhibits high specificity for targeting membrane proteins and inducing efficient lysosomal degradation. SApts were synthesized by conjugating the YXXØ signal peptide to the aptamer's terminus through a click reaction. Our study demonstrated that SApts efficiently degrade disease-associated membrane proteins, such as PTK7, Met, and NCL, based on the inherent signals rather than specific LTR. The potent antitumor efficacy of SApts was further confirmed in a xenograft tumor model, where in vivo degradation of PTK7 was observed. Collectively, our work provides insights into the development of a simple and universal lysosomal degradation platform with potential translational value in clinical treatment.
Collapse
Affiliation(s)
- Wanlin Xie
- School of Materials Science and Engineering, Tianjin University, Tianjin 300350, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Weidi Sun
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Qin Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou 310024, Zhejiang, China
| | - Yang Dang
- School of Materials Science and Engineering, Tianjin University, Tianjin 300350, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Lele Ma
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
| | - Yuan Liu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou 310024, Zhejiang, China
| | - Hui Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou 310024, Zhejiang, China
| | - Fengli Qu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou 310024, Zhejiang, China
| | - Weihong Tan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, Zhejiang, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou 310024, Zhejiang, China
| |
Collapse
|
2
|
Hassane AMA, Obiedallah M, Karimi J, Khattab SMR, Hussein HR, Abo-Dahab Y, Eltoukhy A, Abo-Dahab NF, Abouelela ME. Unravelling fungal genome editing revolution: pathological and biotechnological application aspects. Arch Microbiol 2025; 207:150. [PMID: 40402294 DOI: 10.1007/s00203-025-04360-w] [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: 03/24/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025]
Abstract
Fungi represent a broad and evolutionarily unique group within the eukaryotic domain, characterized by extensive ecological adaptability and metabolic versatility. Their inherent biological intricacy is evident in the diverse and dynamic relationships they establish with various hosts and environmental niches. Notably, fungi are integral to disease processes and a wide array of biotechnological innovations, highlighting their significance in medical, agricultural, and industrial domains. Recent advances in genetic engineering have revolutionized fungal research, with CRISPR/Cas emerging as the most potent and versatile genome editing platform. This technology enables precise manipulation of fungal genomes, from silencing efflux pump genes in Candida albicans (enhancing antifungal susceptibility) to targeting virulence-associated sirtuins in Aspergillus fumigatus (attenuating pathogenicity). Its applications span gene overexpression, multiplexed mutagenesis, and secondary metabolite induction, proving transformative for disease management and biotechnological innovation. CRISPR/Cas9's advantages-unmatched precision, cost-effectiveness, and therapeutic potential-are tempered by challenges like off-target effects, ethical dilemmas, and regulatory gaps. Integrating nanoparticle delivery systems and multi-omics approaches may overcome technical barriers, but responsible innovation requires addressing these limitations. CRISPR-driven fungal genome editing promises to redefine solutions for drug-resistant infections, sustainable bioproduction, and beyond as the field evolves. In conclusion, genome editing technologies have enhanced our capacity to dissect fungal biology and expanded fungi's practical applications across various scientific and industrial domains. Continued innovation in this field promises to unlock the vast potential of fungal systems further, enabling more profound understanding and transformative biotechnological progress.
Collapse
Affiliation(s)
- Abdallah M A Hassane
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt.
| | - Marwa Obiedallah
- Department of Botany and Microbiology, Faculty of Science, University of Sohag, Sohag, 82524, Egypt
| | - Javad Karimi
- Department of Biology, College of Science, Shiraz University, Shiraz, Iran
| | - Sadat M R Khattab
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
- Institute of Advanced Energy, Kyoto University, Gokasho, Uji, Kyoto, 611-0011, Japan
| | - Hussein R Hussein
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Youssef Abo-Dahab
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California, San Francisco 1700 Fourth St, San Francisco, CA, USA
| | - Adel Eltoukhy
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
- Colin Ratledge Center for Microbial Lipids, School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, 255000, China
| | - Nageh F Abo-Dahab
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Mohamed E Abouelela
- Department of Pharmacognosy, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| |
Collapse
|
3
|
Zhang Z, Fan YN, Jiang SQ, Ma YJ, Yu YR, Qing YX, Li QR, Liu YL, Shen S, Wang J. Recent Advances in mRNA Delivery Systems for Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e17571. [PMID: 40391789 DOI: 10.1002/advs.202417571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 04/01/2025] [Indexed: 05/22/2025]
Abstract
mRNA therapy is a promising approach in oncology, offering innovative applications such as tumor vaccines, protein replacement therapy, cell therapy, and gene therapy. However, challenges such as mRNA stability and delivery efficiency must be addressed. Advances in delivery system technologies are crucial for precise mRNA delivery, enhancing treatment safety and efficacy. The development of delivery systems requires accurate organ or cell targeting, intelligent release mechanisms, and optimized administration routes. This review outlines the applications of mRNA therapy in oncology, as well as the utilization of nonviral vectors, encompassing organic, inorganic, and biomimetic systems. It further elucidates the strategies for passive and active vector targeting and examines recent advances in the realm of stimuli-responsive delivery systems that are sensitive to pH and ultrasound. Additionally, the review addresses the development of noninvasive mRNA delivery systems designed for oral and pulmonary administration. The current challenges and emerging trends of mRNA therapy are discussed, and the potential strategies to mitigate these issues are emphasized.
Collapse
Affiliation(s)
- Zheng Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Ya-Nan Fan
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Si-Qi Jiang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Ya-Jing Ma
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Yao-Ru Yu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Yu-Xin Qing
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Qian-Ru Li
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Yi-Lin Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Song Shen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Jun Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| |
Collapse
|
4
|
Han J, Yuan Y, Zhang J, Hou Y, Xu H, Nie X, Zhao Z, Hou J. Regulatory effect of Wnt signaling on mitochondria in cancer: from mechanism to therapy. Apoptosis 2025:10.1007/s10495-025-02114-z. [PMID: 40257508 DOI: 10.1007/s10495-025-02114-z] [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] [Accepted: 04/06/2025] [Indexed: 04/22/2025]
Abstract
Cancer is one of the most significant public health challenges in the new millennium, and complex mechanisms are at work to contribute to its pathogenesis and progression. The Wnt signaling pathways, which are crucial conserved cascades involved in embryological development and tissue homeostasis, and mitochondria, the intracellular powerhouses responsible for energy production, calcium and iron homeostasis, as well as mitochondrial apoptosis in eukaryotic cells, have their own mechanisms regulating these pathological processes. In the past decade, accumulating evidence has indicated that Wnt signaling pathways directly regulate mitochondrial biogenesis and function under physiological and pathological conditions. In this review, we systemically summarize the current understanding of how Wnt signaling pathways, particularly the canonical Wnt cascade, regulate mitochondrial fission, respiration, metabolism, and mitochondrial-dependent apoptosis in cancer. In addition, we discuss recent advancements in the research of anticancer agents and related pharmacological mechanisms targeting the signaling transduction of canonical Wnt pathway and/or mitochondrial function. We believe that the combined use of pharmaceuticals targeting Wnt signaling and/or mitochondria with conventional therapies, immunotherapy and targeted therapy based on accurate molecular pathological diagnosis will undoubtedly be the future mainstream direction of personalized cancer treatment, which could benefit more cancer patients.
Collapse
Affiliation(s)
- Jinping Han
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Yimeng Yuan
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Jianhua Zhang
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd, 475003, Kaifeng, China
| | - Yifan Hou
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Hongtao Xu
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China
| | - Xiaobo Nie
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, 475004, Kaifeng, China.
| | - Zhenhua Zhao
- Ma'anshan 86 Hospital, China RongTong Medical Healthcare Group Co. Ltd, 243100, Ma'anshan, China
| | - Junqing Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd, 475003, Kaifeng, China
| |
Collapse
|
5
|
Han X, Zhang X, Kang L, Feng S, Li Y, Zhao G. Peptide-modified nanoparticles for doxorubicin delivery: Strategies to overcome chemoresistance and perspectives on carbohydrate polymers. Int J Biol Macromol 2025; 299:140143. [PMID: 39855525 DOI: 10.1016/j.ijbiomac.2025.140143] [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: 12/16/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Chemotherapy serves as the primary treatment for cancers, facing challenges due to the emergence of drug resistance. Combination therapy has been developed to combat cancer drug resistance, yet it still suffers from lack of specific targeting of cancer cells and poor accumulation at the tumor site. Consequently, targeted administration of chemotherapy medications has been employed in cancer treatment. Doxorubicin (DOX) is one of the most frequently used chemotherapeutics, functioning by inhibiting topoisomerase activity. Enhancing the anti-cancer effects of DOX and overcoming drug resistance can be accomplished via delivery by nanoparticles. This review will focus on the development of peptide-DOX conjugates, the functionalization of nanoparticles with peptides, the co-delivery of DOX and peptides, as well as the theranostic use of peptide-modified nanoparticles in cancer treatment. The peptide-DOX conjugates have been designed to enhance the targeted delivery to cancer cells by interacting with receptors that are overexpressed on tumor surfaces. Moreover, nanoparticles can be modified with peptides to improve their uptake in tumor cells via endocytosis. Nanoparticles have the ability to co-deliver DOX along with therapeutic peptides for enhanced cancer treatment. Finally, nanoparticles modified with peptides can offer theranostic capabilities by facilitating both imaging and the delivery of DOX (chemotherapy).
Collapse
Affiliation(s)
- Xu Han
- Department of Traditional Chinese medicine, The First Hospital of China Medical University, Shenyang, China
| | - Xue Zhang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, China
| | - Longdan Kang
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Shuai Feng
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, China.
| | - Yinyan Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| | - Ge Zhao
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
6
|
Omidian H, Cubeddu LX, Wilson RL. Peptide-Functionalized Nanomedicine: Advancements in Drug Delivery, Diagnostics, and Biomedical Applications. Molecules 2025; 30:1572. [PMID: 40286158 PMCID: PMC11990896 DOI: 10.3390/molecules30071572] [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: 02/28/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
Peptide-functionalized nanomedicine has emerged as a transformative approach in precision therapeutics and diagnostics, leveraging the specificity of peptides to enhance the performance of nanocarriers, including gold nanoparticles, polymeric nanoparticles, liposomes, mesoporous silica nanoparticles, and quantum dots. These systems enable targeted drug delivery, molecular imaging, biosensing, and regenerative medicine, offering unparalleled advantages in bioavailability, cellular uptake, and therapeutic selectivity. This review provides a comprehensive analysis of peptide-functionalization strategies, nanocarrier design, and their applications across oncology, neurodegenerative disorders, inflammatory diseases, infectious diseases, and tissue engineering. We further discuss the critical role of physicochemical characterization, in vitro and in vivo validation, and regulatory considerations in translating these technologies into clinical practice. Despite the rapid progress in peptide-functionalized platforms, challenges related to stability, immune response, off-target effects, and large-scale reproducibility remain key obstacles to their widespread adoption. Addressing these through advanced peptide engineering, optimized synthesis methodologies, and regulatory harmonization will be essential for their clinical integration. By bridging fundamental research with translational advancements, this review provides an interdisciplinary roadmap for the next generation of peptide-functionalized nanomedicines poised to revolutionize targeted therapy and diagnostics.
Collapse
Affiliation(s)
- Hossein Omidian
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| | - Luigi X. Cubeddu
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| | | |
Collapse
|
7
|
Chen J, Huang Z, Chen Y, Tian H, Chai P, Shen Y, Yao Y, Xu S, Ge S, Jia R. Lactate and lactylation in cancer. Signal Transduct Target Ther 2025; 10:38. [PMID: 39934144 PMCID: PMC11814237 DOI: 10.1038/s41392-024-02082-x] [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: 06/28/2024] [Revised: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 02/13/2025] Open
Abstract
Accumulated evidence has implicated the diverse and substantial influence of lactate on cellular differentiation and fate regulation in physiological and pathological settings, particularly in intricate conditions such as cancer. Specifically, lactate has been demonstrated to be pivotal in molding the tumor microenvironment (TME) through its effects on different cell populations. Within tumor cells, lactate impacts cell signaling pathways, augments the lactate shuttle process, boosts resistance to oxidative stress, and contributes to lactylation. In various cellular populations, the interplay between lactate and immune cells governs processes such as cell differentiation, immune response, immune surveillance, and treatment effectiveness. Furthermore, communication between lactate and stromal/endothelial cells supports basal membrane (BM) remodeling, epithelial-mesenchymal transitions (EMT), metabolic reprogramming, angiogenesis, and drug resistance. Focusing on lactate production and transport, specifically through lactate dehydrogenase (LDH) and monocarboxylate transporters (MCT), has shown promise in the treatment of cancer. Inhibitors targeting LDH and MCT act as both tumor suppressors and enhancers of immunotherapy, leading to a synergistic therapeutic effect when combined with immunotherapy. The review underscores the importance of lactate in tumor progression and provides valuable perspectives on potential therapeutic approaches that target the vulnerability of lactate metabolism, highlighting the Heel of Achilles for cancer treatment.
Collapse
Affiliation(s)
- Jie Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ziyue Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ya Chen
- Department of Radiology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Hao Tian
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Yongning Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Shiqiong Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| |
Collapse
|
8
|
Yang EL, Wang WY, Liu YQ, Yi H, Lei A, Sun ZJ. Tumor-Targeted Catalytic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413210. [PMID: 39676382 DOI: 10.1002/adma.202413210] [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: 09/04/2024] [Revised: 10/30/2024] [Indexed: 12/17/2024]
Abstract
Cancer immunotherapy holds significant promise for improving cancer treatment efficacy; however, the low response rate remains a considerable challenge. To overcome this limitation, advanced catalytic materials offer potential in augmenting catalytic immunotherapy by modulating the immunosuppressive tumor microenvironment (TME) through precise biochemical reactions. Achieving optimal targeting precision and therapeutic efficacy necessitates a thorough understanding of the properties and underlying mechanisms of tumor-targeted catalytic materials. This review provides a comprehensive and systematic overview of recent advancements in tumor-targeted catalytic materials and their critical role in enhancing catalytic immunotherapy. It highlights the types of catalytic reactions, the construction strategies of catalytic materials, and their fundamental mechanisms for tumor targeting, including passive, bioactive, stimuli-responsive, and biomimetic targeting approaches. Furthermore, this review outlines various tumor-specific targeting strategies, encompassing tumor tissue, tumor cell, exogenous stimuli-responsive, TME-responsive, and cellular TME targeting strategies. Finally, the discussion addresses the challenges and future perspectives for transitioning catalytic materials into clinical applications, offering insights that pave the way for next-generation cancer therapies and provide substantial benefits to patients in clinical settings.
Collapse
Affiliation(s)
- En-Li Yang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Wu-Yin Wang
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Ying-Qi Liu
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Hong Yi
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Aiwen Lei
- The Institute for Advanced Studies (IAS), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430079, China
| | - Zhi-Jun Sun
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
9
|
Ren XH, Guo T, Xu MF, Huang Y, Liao XR, Qi LJ, Cheng SX. A Multiple Targeting Genome Editing System for Remodulation of Circulating Malignant Cells to Eliminate Cancer Immunosuppression and Restore Immune Responses. Adv Healthc Mater 2025; 14:e2401223. [PMID: 39440615 DOI: 10.1002/adhm.202401223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/29/2024] [Indexed: 10/25/2024]
Abstract
Cancer immunotherapy, which aims to eliminate cancer immunosuppression and reactivate anticancer immunity, holds great promise in oncology treatments. However, it is challenging to accurately study the efficacy of immunotherapy based on human-derived cells through animal experiments due to xenogeneic immune rejection. Herein, a personalized and precise strategy to evaluate the effectiveness of immunotherapy using the blood samples of cancer patients is presented. Through the utilization of multiple cancer-targeting delivery system decorated with the epidermal growth factor receptor (EGFR)-specific aptamer CL4 and the AXL-specific aptamer GL21.T to achieve superior efficiency in delivering the genome editing plasmid for MUC1 knockout, effective modulation on the behavior of circulating malignant cells (CMCs) is realized. After genome editing, both mucin 1 (MUC1) and programmed death-ligand 1 (PD-L1) are significantly downregulated in CMCs. The elimination of immunosuppression results in markedly enhanced secretion of pro-inflammatory anticancer cytokines encompassing interleukins 2, 12, and 15 and interferon-γ by immune cells. The study not only provides a strategy to overcome immunosuppression but also yields critical insights for personalized immunotherapy approaches.
Collapse
Affiliation(s)
- Xiao-He Ren
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230011, China
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Tao Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, Anhui, 230011, China
| | - Ma-Fei Xu
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230011, China
| | - Yun Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, Anhui, 230011, China
| | - Xin-Ru Liao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Li-Jin Qi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Si-Xue Cheng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| |
Collapse
|
10
|
Hou Y, Zhao Z, Li P, Cao Y, Zhang Y, Guo C, Nie X, Hou J. Combination therapies with Wnt signaling inhibition: A better choice for prostate cancer treatment. Biochim Biophys Acta Rev Cancer 2024; 1879:189186. [PMID: 39332651 DOI: 10.1016/j.bbcan.2024.189186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024]
Abstract
The intractability and high mortality rate of castration-resistant prostate cancer (CRPC) remain the most challenging problems in the field of prostate cancer (PCa). Emerging evidence has shown that the dysregulation of Wnt signaling pathways, which are highly conserved cascades that regulate embryonic development and maintain tissue homeostasis, is involved in various stages of PCa occurrence and progression. In this review, we systemically discuss the mechanisms by which the androgen receptor (AR) signaling pathway and Wnt signaling pathways participate in the occurrence of PCa and its progression to CRPC. Specifically, we elaborate on how Wnt signaling pathways induce the malignant transformation of prostate cells, promote the malignant progression of PCa and establish an immunosuppressive prostate tumor microenvironment through interaction with the AR pathway or in an AR-independent manner. We also discuss how Wnt signaling pathways enhances the stemness characteristics of prostate cancer stem cells (PCSCs) to induce the occurrence and metastasis of CPPC. Additionally, we discuss the latest progress in the use of different types of drugs that inhibit the Wnt signaling pathways in the treatment of PCa. We believe that the combination of Wnt signaling-based drugs with endocrine and other therapies is necessary and may enhance the clinical efficacy in the treatment of all types of PCa.
Collapse
Affiliation(s)
- Yifan Hou
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng 475003, China
| | - Zhenhua Zhao
- Ma'anshan 86 Hospital, China RongTong Medical Healthcare Group Co. Ltd, Ma'anshan 243100, China
| | - Pan Li
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yujia Cao
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yi Zhang
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Changsheng Guo
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng 475003, China
| | - Xiaobo Nie
- Key Laboratory of Receptors-Mediated Gene Regulation, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China.
| | - Junqing Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng 475003, China.
| |
Collapse
|
11
|
Feng X, Li Z, Liu Y, Chen D, Zhou Z. CRISPR/Cas9 technology for advancements in cancer immunotherapy: from uncovering regulatory mechanisms to therapeutic applications. Exp Hematol Oncol 2024; 13:102. [PMID: 39427211 PMCID: PMC11490091 DOI: 10.1186/s40164-024-00570-y] [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: 07/25/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
In recent years, immunotherapy has developed rapidly as a new field of tumour therapy. However, the efficacy of tumour immunotherapy is not satisfactory due to the immune evasion mechanism of tumour cells, induction of immunosuppressive tumour microenvironment (TME), and reduction of antigen delivery, etc. CRISPR/Cas9 gene editing technology can accurately modify immune and tumour cells in tumours, and improve the efficacy of immunotherapy by targeting immune checkpoint molecules and immune regulatory genes, which has led to the great development and application. In current clinical trials, there are still many obstacles to the application of CRISPR/Cas9 in tumour immunotherapy, such as ensuring the accuracy and safety of gene editing, overcoming overreactive immune responses, and solving the challenges of in vivo drug delivery. Here we provide a systematic review on the application of CRISPR/Cas9 in tumour therapy to address the above existing problems. We focus on CRISPR/Cas9 screening and identification of immunomodulatory genes, targeting of immune checkpoint molecules, manipulation of immunomodulators, enhancement of tumour-specific antigen presentation and modulation of immune cell function. Second, we also highlight preclinical studies of CRISPR/Cas9 in animal models and various delivery systems, and evaluate the efficacy and safety of CRISPR/Cas9 technology in tumour immunotherapy. Finally, potential synergistic approaches for combining CRISPR/Cas9 knockdown with other immunotherapies are presented. This study underscores the transformative potential of CRISPR/Cas9 to reshape the landscape of tumour immunotherapy and provide insights into novel therapeutic strategies for cancer patients.
Collapse
Affiliation(s)
- Xiaohang Feng
- Department of Colorectal Surgery, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhengxing Li
- Department of Colorectal Surgery, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuping Liu
- Department of Colorectal Surgery, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Di Chen
- Biomedical Sciences, College of Medicine and Veterinary Medicine, Edinburgh Medical School, The University of Edinburgh, Edinburgh, UK
- Center for Reproductive Medicine of The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhuolong Zhou
- Department of Colorectal Surgery, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Biomedical Sciences, College of Medicine and Veterinary Medicine, Edinburgh Medical School, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
12
|
Li Y, Li C, Yan J, Liao Y, Qin C, Wang L, Huang Y, Yang C, Wang J, Ding X, Yang YY, Yuan P. Polymeric micellar nanoparticles for effective CRISPR/Cas9 genome editing in cancer. Biomaterials 2024; 309:122573. [PMID: 38677222 DOI: 10.1016/j.biomaterials.2024.122573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 03/21/2024] [Accepted: 04/12/2024] [Indexed: 04/29/2024]
Abstract
The clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein 9 (Cas9) gene editing has attracted extensive attentions in various fields, however, its clinical application is hindered by the lack of effective and safe delivery system. Herein, we reported a cationic micelle nanoparticle composed of cholesterol-modified branched small molecular PEI (PEI-CHO) and biodegradable PEG-b-polycarbonate block copolymer (PEG-PC), denoted as PEG-PC/PEI-CHO/pCas9, for the CRISPR/Cas9 delivery to realize genomic editing in cancer. Specifically, PEI-CHO condensed pCas9 into nanocomplexes, which were further encapsulated into PEG-PC nanoparticles (PEG-PC/PEI-CHO/pCas9). PEG-PC/PEI-CHO/pCas9 had a PEG shell, protecting DNA from degradation by nucleases. Enhanced cellular uptake of PEG-PC/PEI-CHO/pCas9 nanoparticles was observed as compared to that mediated by Lipo2k/pCas9 nanoparticles, thus leading to significantly elevated transfection efficiency after escaping from endosomes via the proton sponge effect of PEI. In addition, the presence of PEG shell greatly improved biocompatibility, and significantly enhanced the in vivo tumor retention of pCas9 compared to PEI-CHO/pCas9. Notably, apparent downregulation of GFP expression could be achieved both in vitro and in vivo by using PEG-PC/PEI-CHO/pCas9-sgGFP nanoparticles. Furthermore, PEG-PC/PEI-CHO/pCas9-sgMcl1 induced effective apoptosis and tumor suppression in a HeLa tumor xenograft mouse model by downregulating Mcl1 expression. This work may provide an alternative paradigm for the efficient and safe genome editing in cancer.
Collapse
Affiliation(s)
- Yuzhen Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Chun Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Jiachang Yan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Ying Liao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Chengyuan Qin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Lingyin Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yi Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Chuan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Republic of Singapore
| | - Jianwei Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Xin Ding
- School of Medicine, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Yi Yan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Republic of Singapore.
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
13
|
Lu H, Guan P, Xu S, Han Y, Liu Z. Boosting Cancer Immunotherapy via Reversing PD-L1-Mediated Immunosuppression with a Molecularly Imprinted Lysosomal Nanodegrader. ACS NANO 2024; 18:23553-23565. [PMID: 39137395 DOI: 10.1021/acsnano.4c07416] [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: 08/15/2024]
Abstract
Immune checkpoint blockade therapy has achieved important clinical advances in several types of tumors, particularly via targeting the PD-1/PD-L1 axis. However, existing therapeutic strategies that suppress the PD-1/PD-L1 signal pathway usually experience low treatment efficacy and the risk of causing autoimmune diseases. Herein, we report a cancer cell-targeted molecularly imprinted lysosomal nanodegrader (MILND) for boosting immune checkpoint blockade therapy against tumors. The MILND, imprinted with the N-terminal epitope of PD-L1 as an imprinting template, could specifically target the PD-L1 on tumor cells to promote cellular uptake. This process further induces the transport of PD-L1 into lysosomes for degradation, ultimately resulting in the downregulation of PD-L1 expression levels on tumor cells. As a result, a T cell-mediated immune response in the body was activated via the blockade of the PD-1/PD-L1 signaling pathway, which triggered a durable antitumor efficacy. In vivo experiments demonstrated that the MILND could effectively accumulate in tumor sites and exhibit strong tumor growth suppression efficacy in a xenograft tumor model without obvious side effects. Therefore, the MILND provides not only a promising strategy for boosting cancer immunotherapy but also insights for developing molecular imprinting-empowered nanomedicines.
Collapse
Affiliation(s)
- Haifeng Lu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Peixin Guan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Shuxin Xu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Yanjie Han
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Zhen Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| |
Collapse
|
14
|
Zhao Y, Fei Y, Zhao Y, Li M, Hu Y, Cai K, Yu SH, Luo Z. Biomineralization-Tuned Nanounits Reprogram the Signal Transducer and Activator of Transcription 3 Signaling for Ferroptosis-Immunotherapy in Cancer Stem Cells. ACS NANO 2024; 18:21268-21287. [PMID: 39083438 DOI: 10.1021/acsnano.4c05084] [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: 08/02/2024]
Abstract
Cancer stem cells (CSCs) are promising targets for improving anticancer treatment outcomes while eliminating recurrence, but their treatment remains a major challenge. Here, we report a nanointegrative strategy to realize CSC-targeted ferroptosis-immunotherapy through spatiotemporally controlled reprogramming of STAT3-regulated signaling circuits. Specifically, STAT3 inhibitor niclosamide (Ni) and an experimental ferroptosis drug (1S, 3R)-RSL3 (RSL3) are integrated into hyaluronic acid-modified amorphous calcium phosphate (ACP) nanounits through biomineralization (CaP-PEG-HA@Ni/RSL3), which could be recognized by CD44-overexpressing CSCs and released in a synchronized manner. Ni inhibits the CSC-intrinsic STAT3-PD-L1 axis to stimulate adaptive immunity and enhance interferon gamma (IFNγ) secretion by CD8+ T cells to downregulate SLC7A11 and SLC3A2 for blocking glutathione biosynthesis. Meanwhile, Ni-dependent STAT3 inhibition also upregulates ACSL4 through downstream signaling and IFNγ feedback. These effects cooperate with RSL3-mediated GPX4 deactivation to induce pronounced ferroptosis. Furthermore, CaP-PEG-HA@Ni/RSL3 also impairs the immunosuppressive M2-like tumor-associated macrophages, while Ca2+ ions released from degraded ACP could chelate with lipid peroxides in ferroptotic CSCs to avoid CD8+ T-cell inhibition, thus boosting the effector function of activated CD8+ T cells. This study offers a cooperative ferroptosis-immunotherapeutic approach for the treatment of refractory cancer.
Collapse
Affiliation(s)
- Youbo Zhao
- Center for Tissue Engineering and Stem Cell Research, Key Laboratory for Autoimmune Disease Research, Department of Hepatic-Biliary-Pancreatic Surgery Affiliate Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang 550025, P. R. China
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yang Fei
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yang Zhao
- Division of Nanomaterials and Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Center for Excellence in Nanoscience, Collaborative Innovation Center of Suzhou Nano Science and Technology, Department of Chemistry, Hefei Science Center of CAS, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing 400044, P. R. China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Chongqing University, Chongqing 400044, P. R. China
| | - Shu-Hong Yu
- Division of Nanomaterials and Chemistry, Hefei National Laboratory for Physical Sciences at the Microscale, CAS Center for Excellence in Nanoscience, Collaborative Innovation Center of Suzhou Nano Science and Technology, Department of Chemistry, Hefei Science Center of CAS, University of Science and Technology of China, Hefei 230026, P. R. China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
15
|
Khademi Z, Yazdi KS, Ramezani M, Alibolandi M, Rezvani SA, Abnous K, Taghdisi SM. FOXM1 Aptamer-Polyethylenimine Nanoplatform Coated With Hyaluronic Acid And AS1411 Aptamer For Dual-Targeted Delivery of Doxorubicin And Synergistic Treatment of Tumor Cells. J Pharm Sci 2024; 113:2198-2207. [PMID: 38432623 DOI: 10.1016/j.xphs.2024.02.025] [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: 01/14/2024] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 03/05/2024]
Abstract
The objective of this investigation was to develop a self-assembled, dual-functionalized delivery system that could effectively transport doxorubicin (DOX) to cancer cells through the use of AS1411 aptamer and hyaluronic acid polymer (HA). The ultimate goal is an improved targeting approach for more efficient treatment. The core of this system comprised polyethylenimine (PEI) and FOXM1 aptamer, which was coated by HA. Next, nucleolin targeting aptamers (AS1411) were loaded onto the nanocomplex. Afterward, DOX was added to Aptamers (Apts)-HA-PEI-FOXM1 NPs to create the DOX-AS1411-HA-PEI-FOXM1 NPs for better treatment of cancer cells. The cytotoxic effect of the nanocomplex on L929, 4T1, and A549 cells showed that cell mortality in target cancer cells (4T1 and A549) was considerably enhanced compared to nontarget cells (L929, normal cells). The findings from the flow cytometry analysis and fluorescence imaging demonstrated the cellular absorption of DOX-Apts-HA-PEI-FOXM1 NPs in target cells was significantly enhanced when compared to L929 cells. Furthermore, in vivo antitumor study exhibited that DOX-Apts-HA-PEI-FOXM1 NPs rendered specific tumor accumulation and increasing of the anti-tumor effects.
Collapse
Affiliation(s)
- Zahra Khademi
- Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Katayoon Sarafraz Yazdi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyede Aysa Rezvani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
16
|
Ilieş BD, Yildiz I, Abbas M. Peptide-conjugated Nanoparticle Platforms for Targeted Delivery, Imaging, and Biosensing Applications. Chembiochem 2024; 25:e202300867. [PMID: 38551557 DOI: 10.1002/cbic.202300867] [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: 12/25/2023] [Revised: 03/09/2024] [Indexed: 04/24/2024]
Abstract
Peptides have become an indispensable tool in engineering of multifunctional nanostructure platforms for biomedical applications such as targeted drug and gene delivery, imaging and biosensing. They can be covalently incorporated into a variety of nanoparticles (NPs) including polymers, metallic nanoparticles, and others. Using different bioconjugation techniques, multifunctional peptide-modified NPs can be formulated to produce therapeutical and diagnostic platforms offering high specificity, lower toxicity, biocompatibility, and stimuli responsive behavior. Targeting peptides can direct the nanoparticles into specific tissues for targeted drug and gene delivery and imaging applications due to their specificity towards certain receptors. Furthermore, due to their stimuli-responsive features, they can offer controlled release of therapeutics into desired sites of disease. In addition, peptide-based biosensors and imaging agents can provide non-invasive detection and monitoring of diseases including cancer, infectious diseases, and neurological disorders. In this review, we covered the design and formulation of recent peptide-based NP platforms, as well as their utilization in in vitro and in vivo applications such as targeted drug and gene delivery, targeting, sensing, and imaging applications. In the end, we provided the future outlook to design new peptide conjugated nanomaterials for biomedical applications.
Collapse
Affiliation(s)
- Bogdan Dragoş Ilieş
- Department of Chemistry, Khalifa University of Science and Technology, P.O. Box, 127788, Abu Dhabi, UAE
- Functional Biomaterials Group, Khalifa University of Science and Technology, P.O. Box, 127788, Abu Dhabi, UAE
| | - Ibrahim Yildiz
- Department of Chemistry, Khalifa University of Science and Technology, P.O. Box, 127788, Abu Dhabi, UAE
- Functional Biomaterials Group, Khalifa University of Science and Technology, P.O. Box, 127788, Abu Dhabi, UAE
| | - Manzar Abbas
- Department of Chemistry, Khalifa University of Science and Technology, P.O. Box, 127788, Abu Dhabi, UAE
- Functional Biomaterials Group, Khalifa University of Science and Technology, P.O. Box, 127788, Abu Dhabi, UAE
| |
Collapse
|
17
|
Mahmoudian F, Ahmari A, Shabani S, Sadeghi B, Fahimirad S, Fattahi F. Aptamers as an approach to targeted cancer therapy. Cancer Cell Int 2024; 24:108. [PMID: 38493153 PMCID: PMC10943855 DOI: 10.1186/s12935-024-03295-4] [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: 11/28/2023] [Accepted: 03/06/2024] [Indexed: 03/18/2024] Open
Abstract
Conventional cancer treatments can cause serious side effects because they are not specific to cancer cells and can damage healthy cells. Aptamers often are single-stranded oligonucleotides arranged in a unique architecture, allowing them to bind specifically to target sites. This feature makes them an ideal choice for targeted therapeutics. They are typically produced through the systematic evolution of ligands by exponential enrichment (SELEX) and undergo extensive pharmacological revision to modify their affinity, specificity, and therapeutic half-life. Aptamers can act as drugs themselves, directly inhibiting tumor cells. Alternatively, they can be used in targeted drug delivery systems to transport drugs directly to tumor cells, minimizing toxicity to healthy cells. In this review, we will discuss the latest and most advanced approaches to using aptamers for cancer treatment, particularly targeted therapy overcoming resistance to conventional therapies.
Collapse
Affiliation(s)
- Fatemeh Mahmoudian
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Azin Ahmari
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
- Department of Radiation Oncology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Shiva Shabani
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
- Department of Infectious Diseases, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Bahman Sadeghi
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
- Department of Community Medicine, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Shohreh Fahimirad
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran.
| | - Fahimeh Fattahi
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Zhou Z, Wang H, Li J, Jiang X, Li Z, Shen J. Recent progress, perspectives, and issues of engineered PD-L1 regulation nano-system to better cure tumor: A review. Int J Biol Macromol 2024; 254:127911. [PMID: 37939766 DOI: 10.1016/j.ijbiomac.2023.127911] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 10/29/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Currently, immune checkpoint blockade (ICB) therapies that target the programmed cell death ligand-1 (PD-L1) have been used as revolutionary cancer treatments in the clinic. Apart from restoring the antitumor response of cytotoxic T cells by blocking the interaction between PD-L1 on tumor cells and programmed cell death-1 (PD-1) on T cells, PD-L1 proteins were also newly revealed to possess the capacity to accelerate DNA damage repair (DDR) and enhance tumor growth through multiple mechanisms, leading to the impaired efficacy of tumor therapies. Nevertheless, current free anti-PD-1/PD-L1 therapy still suffered from poor therapeutic outcomes in most solid tumors due to the non-selective tumor accumulation, ineludible severe cytotoxic effects, as well as the common occurrence of immune resistance. Recently, nanoparticles with efficient tumor-targeting capacity, tumor-responsive prosperity, and versatility for combination therapy were identified as new avenues for PD-L1 targeting cancer immunotherapies. In this review, we first summarized the multiple functions of PD-L1 protein in promoting tumor growth, accelerating DDR, as well as depressing immunotherapy efficacy. Following this, the effects and mechanisms of current clinically widespread tumor therapies on tumor PD-L1 expression were discussed. Then, we reviewed the recent advances in nanoparticles for anti-PD-L1 therapy via using PD-L1 antibodies, small interfering RNA (siRNA), microRNA (miRNA), clustered, regularly interspaced, short palindromic repeats (CRISPR), peptide, and small molecular drugs. At last, we discussed the challenges and perspectives to promote the clinical application of nanoparticles-based PD-L1-targeting therapy.
Collapse
Affiliation(s)
- Zaigang Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Haoxiang Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jie Li
- College of Pharmacy, Wenzhou Medical University, Wenzhou 325000, China
| | - Xin Jiang
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhangping Li
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang 324000, China.
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China.
| |
Collapse
|
19
|
Yuan S, Che Y, Wang Z, Xing K, Xie X, Chen Y. Mitochondrion-targeted carboxymethyl chitosan hybrid nanoparticles loaded with Coenzyme Q10 protect cardiac grafts against cold ischaemia‒reperfusion injury in heart transplantation. J Transl Med 2023; 21:925. [PMID: 38124174 PMCID: PMC10734076 DOI: 10.1186/s12967-023-04763-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Heart transplantation (HT) has been approved as an optimal therapeutic regimen for patients with terminal-stage cardiac failure. However, cold ischaemia‒reperfusion (I/R) injury remains an unavoidable and outstanding challenge, which is a major factor in early graft dysfunction and an obstacle to long-term survival in HT. Cold I/R injury induces cardiac graft injury by promoting mitochondrial dysfunction and augmenting free radical production and inflammatory responses. We therefore designed a mitochondrion-targeted nanocarrier loaded with Coenzyme Q10 (CoQ10) (CoQ10@TNPs) for treatment of cold I/R injury after cardiac graft in a murine heterotopic cardiac transplantation model. METHODS Hybrid nanoparticles composed of CaCO3/CaP/biotinylated-carboxymethylchitosan (CaCO3/CaP/BCMC) were synthesized using the coprecipitation method, and the mitochondria-targeting tetrapeptide SS31 was incorporated onto the surface of the hybrid nanoparticles through biotin-avidin interactions. Transmission electron microscopy (TEM) and dynamic light scattering (DLS) analysis were used for characterisation. In vitro, the hypoxia-reoxygenation model of H9c2 cells was employed to replicate in vivo cold I/R injury and treated with CoQ10@TNPs. The impact of CoQ10@TNPs on H9c2 cell injury was assessed by analysis of oxidative damage and apoptosis. In vivo, donor hearts (DHs) were perfused with preservation solution containing CoQ10@TNPs and stored in vitro at 4 °C for 12 h. The DHs were heterotopically transplanted and analysed for graft function, oxidative damage, apoptosis, and inflammatory markers 1 day post-transplantation. RESULTS CoQ10@TNPs were successfully synthesized and delivered CoQ10 to the mitochondria of the cold ischaemic myocardium. In vitro experiments demonstrated that CoQ10@TNPs was taken up by H9c2 cells at 4 °C and localized within the mitochondria, thus ameliorating oxidative stress damage and mitochondrial injury in cold I/R injury. In vivo experiments showed that CoQ10@TNPs accumulated in DH tissue at 4 °C, localized within the mitochondria during cold storage and improved cardiac graft function by attenuating mitochondrial oxidative injury and inflammation. CONCLUSIONS CoQ10@TNPs can precisely deliver CoQ10 to the mitochondria of cold I/R-injured cardiomyocytes to effectively eliminate mitochondrial reactive oxygen species (mtROS), thus reducing oxidative injury and inflammatory reactions in cold I/R-injured graft tissues and finally improving heart graft function. Thus, CoQ10@TNPs offer an effective approach for safeguarding cardiac grafts against extended periods of cold ischaemia, emphasizing the therapeutic potential in mitigating cold I/R injury during HT. These findings present an opportunity to enhance existing results following HT and broaden the range of viable grafts for transplantation.
Collapse
Affiliation(s)
- Shun Yuan
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan, 430000, Hubei, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanjia Che
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan, 430000, Hubei, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, Hubei, People's Republic of China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan, 430000, Hubei, People's Republic of China.
| | - Kai Xing
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan, 430000, Hubei, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoping Xie
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan, 430000, Hubei, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuanyang Chen
- Department of Cardiovascular Surgery, Renmin Hospital of Wuhan University, 238# Jiefang Road, Wuhan, 430000, Hubei, People's Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
20
|
Fu L, Qi C, Sun T, Huang K, Lin J, Huang P. Glucose oxidase-instructed biomineralization of calcium-based biomaterials for biomedical applications. EXPLORATION (BEIJING, CHINA) 2023; 3:20210110. [PMID: 38264686 PMCID: PMC10742215 DOI: 10.1002/exp.20210110] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 02/22/2023] [Indexed: 01/25/2024]
Abstract
In recent years, glucose oxidase (GOx) has aroused great research interest in the treatment of diseases related to abnormal glucose metabolisms like cancer and diabetes. However, as a kind of endogenous oxido-reductase, GOx suffers from poor stability and system toxicity in vivo. In order to overcome this bottleneck, GOx is encapsulated in calcium-based biomaterials (CaXs) such as calcium phosphate (CaP) and calcium carbonate (CaCO3) by using it as a biotemplate to simulate the natural biomineralization process. The biomineralized GOx holds improved stability and reduced side effects, due to the excellent bioactivity, biocompatibitliy, and biodegradability of CaXs. In this review, the state-of-the-art studies on GOx-mineralized CaXs are introduced with an emphasis on their application in various biomedical fields including disease diagnosis, cancer treatment, and diabetes management. The current challenges and future perspectives of GOx-mineralized CaXs are discussed, which is expected to promote further studies on these smart GOx-mineralized CaXs biomaterials for practical applications.
Collapse
Affiliation(s)
- Lian‐Hua Fu
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Laboratory of Evolutionary Theranostics (LET)School of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhenChina
| | - Chao Qi
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Laboratory of Evolutionary Theranostics (LET)School of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhenChina
| | - Tuanwei Sun
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Laboratory of Evolutionary Theranostics (LET)School of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhenChina
| | - Kai Huang
- Department of Materials Science and EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Jing Lin
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Laboratory of Evolutionary Theranostics (LET)School of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhenChina
| | - Peng Huang
- Marshall Laboratory of Biomedical EngineeringInternational Cancer Center, Laboratory of Evolutionary Theranostics (LET)School of Biomedical EngineeringShenzhen University Medical SchoolShenzhen UniversityShenzhenChina
| |
Collapse
|
21
|
Chowdhry R, Lu SZ, Lee S, Godhulayyagari S, Ebrahimi SB, Samanta D. Enhancing CRISPR/Cas systems with nanotechnology. Trends Biotechnol 2023; 41:1549-1564. [PMID: 37451945 DOI: 10.1016/j.tibtech.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
CRISPR/Cas systems have revolutionized biology and medicine, and have led to new paradigms in disease diagnostics and therapeutics. However, these complexes suffer from key limitations regarding barriers to cellular entry, stability in biological environments, and off-target effects. Integrating nanotechnology with CRISPR/Cas systems has emerged as a promising strategy to overcome these challenges and has further unlocked structures that accumulate preferentially in tissues of interest, have tunable pharmacological properties, and are activated in response to desired stimuli. Nanomaterials can also enhance CRISPR/Cas-mediated detection platforms by enabling faster, more sensitive, and convenient readouts. We highlight recent advances in this rapidly growing field. We also outline areas that need further development to fully realize the potential of CRISPR technologies.
Collapse
Affiliation(s)
- Rupali Chowdhry
- Department of Public Health, The University of Texas at Austin, Austin, TX 78712, USA
| | - Steven Z Lu
- Department of Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Seungheon Lee
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA
| | | | - Sasha B Ebrahimi
- Drug Product Development - Steriles, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - Devleena Samanta
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
22
|
Han D, Ren XH, He XY, Chen XS, Pang X, Cheng SX. Aptamer/Peptide-Functionalized Nanoprobe for Detecting Multiple miRNAs in Circulating Malignant Cells to Study Tumor Heterogeneity. ACS Biomater Sci Eng 2023; 9:5832-5842. [PMID: 37679307 DOI: 10.1021/acsbiomaterials.3c01055] [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: 09/09/2023]
Abstract
Identification of diverse biomarkers in heterogenic circulating malignant cells (CMCs) such as circulating tumor cells (CTCs) and circulating tumor endothelial cells (CTECs) has crucial significance in tumor diagnosis. However, it remains a substantial challenge to achieve in situ detection of multiple miRNA markers in living cells in blood. Herein, we demonstrate that an aptamer/peptide-functionalized vector can deliver molecular beacons into targeted living CMCs in peripheral blood of patients for in situ detection of multiple cancer biomarkers, including miRNA-21 (miR-21) and miRNA-221 (miR-221). Based on miR-21 and miR-221 levels, heterogenic CMCs are identified for both nondistant metastatic and distant metastatic cancer patients. CMCs from nondistant metastatic and distant metastatic cancer patients exhibit similar miR-21 levels, while the miR-221 level in CMCs of the distant metastatic cancer patient is higher than that of the nondistant metastatic cancer patient. With the capability to realize precise probing of multiple intracellular biomarkers in living CMCs at the single-cell resolution, the nanoprobe can reveal the tumor heterogeneity and provide useful information for diagnosis and prognosis. The nanoprobe we developed would accelerate the progress toward noninvasive precise cancer diagnosis.
Collapse
Affiliation(s)
- Di Han
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, China
- School of Life Sciences and Technology, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xiao-He Ren
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Xiao-Yan He
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Xue-Si Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xuan Pang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
23
|
Chen KH, Nguyen N, Huang TY, Lin YJ, Yu YT, Song HL, Wang JT, Nguyen VK, Chen HL, Chu LA, Chiang HHK, Sung HW. Macrophage-Hitchhiked Orally Administered β-Glucans-Functionalized Nanoparticles as "Precision-Guided Stealth Missiles" for Targeted Pancreatic Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304735. [PMID: 37363886 DOI: 10.1002/adma.202304735] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/23/2023] [Indexed: 06/28/2023]
Abstract
The prognosis in cases of pancreatic ductal adenocarcinoma (PDAC) with current treatment modalities is poor owing to the highly desmoplastic tumor microenvironment (TME). Herein, a β-glucans-functionalized zinc-doxorubicin nanoparticle system (βGlus-ZnD NPs) that can be orally administered, is developed for targeted PDAC therapy. Following oral administration in PDAC-bearing mice, βGlus-ZnD NPs actively target/transpass microfold cells, overcome the intestinal epithelial barrier, and then undergo subsequent phagocytosis by endogenous macrophages (βGlus-ZnD@Mϕ). As hitchhiking cellular vehicles, βGlus-ZnD@Mϕ transits through the intestinal lymphatic system and enters systemic circulation, ultimately accumulating in the tumor tissue as a result of the tumor-homing and "stealth" properties that are conferred by endogenous Mϕ. Meanwhile, the Mϕ that hitchhikes βGlus-ZnD NPs is activated to produce matrix metalloproteinases, destroying the desmoplastic stromal barrier, and differentiates toward the M1 -like phenotype, modulating the TME and recruiting effector T cells, ultimately inducing apoptosis of the tumor cells. The combination of βGlus-ZnD@Mϕ and immune checkpoint blockade effectively inhibits the growth of the primary tumor and suppresses the development of metastasis. It thus represents an appealing approach to targeted PDAC therapy.
Collapse
Affiliation(s)
- Kuan-Hung Chen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Nhien Nguyen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Tun-Yu Huang
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Yu-Jung Lin
- Research Center for Applied Sciences, 11529, Academia Sinica, Taipei, Taiwan
| | - Yu-Tzu Yu
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Hsiang-Lin Song
- Department of Pathology, National Taiwan University Hospital, 300, Hsinchu Branch, Hsinchu, Taiwan
| | - Jui-To Wang
- Neurological Institute, Department of Neurosurgery, Taipei Veterans General Hospital, 11217, Taipei, Taiwan
- Institute of Brain Science, National Yang-Ming Chiao Tung University, 11221, Taipei, Taiwan
| | - Van Khanh Nguyen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Hsin-Lung Chen
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Li-An Chu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 30013, Hsinchu, Taiwan
| | - Hui-Hua Kenny Chiang
- Institute of Biomedical Engineering, National Yang-Ming Chiao Tung University, 11221, Taipei, Taiwan
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 30013, Hsinchu, Taiwan
| |
Collapse
|
24
|
Xiang J, Liu K, Xu H, Zhao Z, Piao Y, Shao S, Tang J, Shen Y, Zhou Z. Dual Synergistic Tumor-Specific Polymeric Nanoparticles for Efficient Chemo-Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301216. [PMID: 37551065 PMCID: PMC10582463 DOI: 10.1002/advs.202301216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/02/2023] [Indexed: 08/09/2023]
Abstract
Chemo-immunotherapy has made significant progress in cancer treatment. However, the cancer cell self-defense mechanisms, including cell cycle checkpoint and programmed cell death-ligand 1 (PD-L1) upregulation, have greatly hindered the therapeutic efficacy. Herein, norcantharidin (NCTD)-platinum (Pt) codelivery nanoparticles (NC-NP) with tumor-sensitive release profiles are designed to overcome the self-defense mechanisms via synergistic chemo-immunotherapy. NC-NP remains stable under normal physiological conditions but quickly releases 1,2-diaminocyclohexane-platinum(II) (DACHPt, a parent drug of oxaliplatin) and NCTD in response to the tumor acidity. NCTD inhibits protein phosphatase 2A (PP2A) activity to relieve cell cycle arrest and downregulates the tumor PD-L1 expression to disrupt the programmed cell death-1 (PD-1)/PD-L1 interaction, synergistically enhancing Pt-based chemotherapy and immunogenic cell death-induced immunotherapy. As a result, NC-NP exhibits potent synergistic cytotoxicity and promotes T cell recruitment to generate robust antitumor immune responses. The dual synergism exhibits potent antitumor activity against orthotopic 4T1 tumors, providing a promising chemo-immunotherapy paradigm for cancer treatment.
Collapse
Affiliation(s)
- Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
- ZJU‐Hangzhou Global Scientific and Technological Innovation CenterZhejiang UniversityHangzhou311215China
| | - Kexin Liu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
| | - Hongxia Xu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
| | - Zhihao Zhao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
- ZJU‐Hangzhou Global Scientific and Technological Innovation CenterZhejiang UniversityHangzhou311215China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
- ZJU‐Hangzhou Global Scientific and Technological Innovation CenterZhejiang UniversityHangzhou311215China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of EducationCollege of Chemical and Biological EngineeringZhejiang UniversityHangzhou310027China
| |
Collapse
|
25
|
Han D, He X, Huang Y, Gao M, Guo T, Ren X, Liao X, Chen X, Pang X, Cheng S. A Multifunctional Delivery System for Remodulating Cell Behaviors of Circulating Malignant Cells to Prevent Cell Fusion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303309. [PMID: 37590231 PMCID: PMC10582411 DOI: 10.1002/advs.202303309] [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: 06/19/2023] [Revised: 07/30/2023] [Indexed: 08/19/2023]
Abstract
Cell fusion plays a critical role in cancer progression and metastasis. However, effective modulation of the cell fusion behavior and timely evaluation on the cell fusion to provide accurate information for personalized therapy are facing challenges. Here, it demonstrates that the cancer cell fusion behavior can be efficiently modulated and precisely detected through employing a multifunctional delivery vector to realize cancer targeting delivery of a genome editing plasmid and a molecular beacon-based AND logic gate. The multifunctional delivery vector decorated by AS1411 conjugated hyaluronic acid and NLS-GE11 peptide conjugated hyaluronic acid can specifically target circulating malignant cells (CMCs) of cancer patients to deliver the genome editing plasmid for epidermal growth factor receptor (EGFR) knockout. The cell fusion between CMCs and endothelial cells can be detected by the AND logic gate delivered by the multifunctional vector. After EGFR knockout, the edited CMCs exhibit dramatically inhibited cell fusion capability, while unedited CMCs can easily fuse with human umbilical vein endothelial cells (HUVEC) to form hybrid cells. This study provides a new therapeutic strategy for preventing cancer progression and a reliable tool for evaluating cancer cell fusion for precise personalized therapy.
Collapse
Affiliation(s)
- Di Han
- Key Laboratory of Biomedical Polymers of Ministry of EducationDepartment of ChemistryWuhan UniversityWuhanHubei430072China
| | - Xiao‐Yan He
- School of Life SciencesAnhui Medical UniversityHefeiAnhui230011China
| | - Yun Huang
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityAnhui Public Health Clinical CenterHefeiAnhui230011China
| | - Min Gao
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Anhui Medical UniversityAnhui Public Health Clinical CenterHefeiAnhui230011China
| | - Tao Guo
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Anhui Medical UniversityAnhui Public Health Clinical CenterHefeiAnhui230011China
| | - Xiao‐He Ren
- Key Laboratory of Biomedical Polymers of Ministry of EducationDepartment of ChemistryWuhan UniversityWuhanHubei430072China
| | - Xin‐Ru Liao
- Key Laboratory of Biomedical Polymers of Ministry of EducationDepartment of ChemistryWuhan UniversityWuhanHubei430072China
| | - Xue‐Si Chen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
| | - Xuan Pang
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
| | - Si‐Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of EducationDepartment of ChemistryWuhan UniversityWuhanHubei430072China
| |
Collapse
|
26
|
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
|
27
|
Bertrand P. Aptamers Targeting the PD-1/PD-L1 Axis: A Perspective. J Med Chem 2023; 66:10878-10888. [PMID: 37561598 DOI: 10.1021/acs.jmedchem.3c00551] [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: 08/12/2023]
Abstract
Aptamers have emerged in recent years as alternatives to antibodies or small molecules to interfere with the immune check points by blocking the PD-1/PD-L1 interactions and represent an interesting perspective for immuno-oncology. Aptamers are RNA or DNA nucleotides able to bind to a target with high affinity, with the target ranging from small molecules to proteins and up to cells. Aptamers are identified by the SELEX method that can be modified for specific purposes. The range of applications of aptamers covers therapy as well as new alternative assay technologies similar to ELISA. Aptamers' limited plasma stability can be managed using delivery strategies. The goal of this Perspective is to give an overview of the current development of aptamers targeting the most studied immune checkpoint modulators, PD-1 and PD-L1, and analogous strategies with aptamers for other immuno-related targets.
Collapse
Affiliation(s)
- Philippe Bertrand
- University of Poitiers, IC2MP UMR 7285 CNRS, 4 rue Michel Brunet B27, TSA 51106, 86073 Poitiers cedex 9, France
| |
Collapse
|
28
|
Liang Y, Zhang J, Xu C, Wang J, Han W, Yang J, Wu S, An J, Liu J, Zhang Z, Shi J, Zhang K. Biomimetic Mineralized CRISPR/Cas RNA Nanoparticles for Efficient Tumor-Specific Multiplex Gene Editing. ACS NANO 2023; 17:15025-15043. [PMID: 37481734 DOI: 10.1021/acsnano.3c04116] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
CRISPR/Cas9 systems have great potential to achieve sophisticated gene therapy and cell engineering by editing multiple genomic loci. However, to achieve efficient multiplex gene editing, the delivery system needs adequate capacity to transfect all CRISPR/Cas9 RNA species at the required stoichiometry into the cytosol of each individual cell. Herein, inspired by biomineralization in nature, we develop an all-in-one biomimetic mineralized CRISPR/Cas9 RNA delivery system. This system allows for precise control over the coencapsulation ratio between Cas9 mRNA and multiple sgRNAs, while also exhibiting a high RNA loading capacity. In addition, it enhances the storage stability of RNA at 4 °C for up to one month, and the surface of the nanoparticles can be easily functionalized for precise targeting of RNA nanoparticles in vivo at nonliver sites. Based on the above characteristics, as a proof-of-concept, our system was able to achieve significant gene-editing at each target gene (Survivin: 31.9%, PLK1: 24.41%, HPV: 23.2%) and promote apoptosis of HeLa cells in the mouse model, inhibiting tumor growth without obvious off-target effects in liver tissue. This system addresses various challenges associated with multicomponent RNA delivery in vivo, providing an innovative strategy for the RNA-based CRISPR/Cas9 gene editing.
Collapse
Affiliation(s)
- Yan Liang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Jingge Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Chenlu Xu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Jinjin Wang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Wenshuai Han
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Jiali Yang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Sixuan Wu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Jingyi An
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, P. R. China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou 450001, P. R. China
| |
Collapse
|
29
|
Li Y, Wei Y, Huang Y, Qin G, Zhao C, Ren J, Qu X. Lactate-Responsive Gene Editing to Synergistically Enhance Macrophage-Mediated Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301519. [PMID: 37156740 DOI: 10.1002/smll.202301519] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/19/2023] [Indexed: 05/10/2023]
Abstract
Combination therapies involving metabolic regulation and immune checkpoint blockade are considered an encouraging new strategy for cancer therapy. However, the effective utilization of combination therapies for activating tumor-associated macrophages (TAMs) remains challenging. Herein, a lactate-catalyzed chemodynamic approach to activate the therapeutic genome editing of signal-regulatory protein α (SIRPα) to reprogram TAMs and improve cancer immunotherapy is proposed. This system is constructed by encapsulating lactate oxidase (LOx) and clustered regularly interspaced short palindromic repeat-mediated SIRPα genome-editing plasmids in a metal-organic framework (MOF). The genome-editing system is released and activated by acidic pyruvate, which is produced by the LOx-catalyzed oxidation of lactate. The synergy between lactate exhaustion and SIRPα signal blockade can enhance the phagocytic ability of TAMs and promote the repolarization of TAMs to the antitumorigenic M1 phenotype. Lactate exhaustion-induced CD47-SIRPα blockade efficiently improves macrophage antitumor immune responses and effectively reverses the immunosuppressive tumor microenvironment to inhibit tumor growth, as demonstrated by in vitro and in vivo studies. This study provides a facile strategy for engineering TAMs in situ by combining CRISPR-mediated SIRPα knockout with lactate exhaustion for effective immunotherapy.
Collapse
Affiliation(s)
- Yuwei Li
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Yue Wei
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Ying Huang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Geng Qin
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Chuanqi Zhao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 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, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 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, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| |
Collapse
|
30
|
Sahel DK, Vora LK, Saraswat A, Sharma S, Monpara J, D'Souza AA, Mishra D, Tryphena KP, Kawakita S, Khan S, Azhar M, Khatri DK, Patel K, Singh Thakur RR. CRISPR/Cas9 Genome Editing for Tissue-Specific In Vivo Targeting: Nanomaterials and Translational Perspective. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207512. [PMID: 37166046 PMCID: PMC10323670 DOI: 10.1002/advs.202207512] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 04/15/2023] [Indexed: 05/12/2023]
Abstract
Clustered randomly interspaced short palindromic repeats (CRISPRs) and its associated endonuclease protein, i.e., Cas9, have been discovered as an immune system in bacteria and archaea; nevertheless, they are now being adopted as mainstream biotechnological/molecular scissors that can modulate ample genetic and nongenetic diseases via insertion/deletion, epigenome editing, messenger RNA editing, CRISPR interference, etc. Many Food and Drug Administration-approved and ongoing clinical trials on CRISPR adopt ex vivo strategies, wherein the gene editing is performed ex vivo, followed by reimplantation to the patients. However, the in vivo delivery of the CRISPR components is still under preclinical surveillance. This review has summarized the nonviral nanodelivery strategies for gene editing using CRISPR/Cas9 and its recent advancements, strategic points of view, challenges, and future aspects for tissue-specific in vivo delivery of CRISPR/Cas9 components using nanomaterials.
Collapse
Affiliation(s)
- Deepak Kumar Sahel
- Department of PharmacyBirla Institute of Technology and Science‐PilaniBITS‐Pilani, Vidya ViharPilaniRajasthan333031India
| | - Lalitkumar K. Vora
- School of PharmacyQueen's University Belfast97 Lisburn RoadBelfastBT9 7BLUK
| | - Aishwarya Saraswat
- College of Pharmacy & Health SciencesSt. John's UniversityQueensNY11439USA
| | - Saurabh Sharma
- Terasaki Institute for Biomedical InnovationLos AngelesCA90064USA
| | - Jasmin Monpara
- Department of Pharmaceutical SciencesUniversity of SciencesPhiladelphiaPA19104USA
| | - Anisha A. D'Souza
- Graduate School of Pharmaceutical Sciences and School of PharmacyDuquesne UniversityPittsburghPA15282USA
| | - Deepakkumar Mishra
- School of PharmacyQueen's University Belfast97 Lisburn RoadBelfastBT9 7BLUK
| | - Kamatham Pushpa Tryphena
- Molecular and Cellular Neuroscience LabDepartment of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)‐HyderabadTelangana500037India
| | - Satoru Kawakita
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCA95616USA
| | - Shahid Khan
- Terasaki Institute for Biomedical InnovationLos AngelesCA90064USA
| | - Mohd Azhar
- Research and Development Tata Medical and Diagnostics LimitedMumbaiMaharashtra400001India
| | - Dharmendra Kumar Khatri
- Molecular and Cellular Neuroscience LabDepartment of Pharmacology and ToxicologyNational Institute of Pharmaceutical Education and Research (NIPER)‐HyderabadTelangana500037India
| | - Ketan Patel
- College of Pharmacy & Health SciencesSt. John's UniversityQueensNY11439USA
| | | |
Collapse
|
31
|
Ma L, Yang S, Peng Q, Zhang J, Zhang J. CRISPR/Cas9-based gene-editing technology for sickle cell disease. Gene 2023; 874:147480. [PMID: 37182559 DOI: 10.1016/j.gene.2023.147480] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/16/2023]
Abstract
Sickle cell disease (SCD) is the most common monogenic hematologic disorder and is essentially congenital hemolytic anemia caused by an inherited point mutation in the β-globin on chromosome 11. Although the genetic basis of SCD was revealed as early as 1957, treatment options for SCD have been very limited to date. Hematopoietic stem cell transplantation (HSCT) was thought to hold promise as a cure for SCD, but the available donors were still only 15% useful. Gene therapy has advanced rapidly into the 21st century with the promise of a cure for SCD, and gene editing strategies based on the cluster-based regularly interspaced short palindromic repeat sequence (CRISPR)/Cas9 system have revolutionized the field of gene therapy by precisely targeting genes. In this paper, we review the pathogenesis and therapeutic approaches of SCD, briefly summarize the delivery strategies of CRISPR/Cas9, and finally discuss in depth the current status, application barriers, and solution directions of CRISPR/Cas9 in SCD. Through the review in this paper, we hope to provide some references for gene therapy in SCD.
Collapse
Affiliation(s)
- Liangliang Ma
- Department of Hematology, Meishan City People's Hospital, Meishan City, Sichuan Province 620000, China
| | - Shanglun Yang
- Department of Hematology, Meishan City People's Hospital, Meishan City, Sichuan Province 620000, China
| | - Qianya Peng
- Department of Hematology, Meishan City People's Hospital, Meishan City, Sichuan Province 620000, China
| | - Jingping Zhang
- Department of Hematology, Meishan City People's Hospital, Meishan City, Sichuan Province 620000, China
| | - Jing Zhang
- Department of Hematology, Meishan City People's Hospital, Meishan City, Sichuan Province 620000, China.
| |
Collapse
|
32
|
Qiao L, Gao M, Yi X, Peng H, Zhang R, Yao W, Sun G, He X. Biomimetic gene editing system for precise tumor cell reprogramming and augmented tumor therapy. J Control Release 2023; 356:663-677. [PMID: 36924897 DOI: 10.1016/j.jconrel.2023.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/22/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
The abnormal level of hypoxia-inducible factor-1 alpha (HIF-1α) is closely related to cancer metastasis and treatment resistance. CRISPR-Cas9-based gene editing technology has sparked profound hope to solve this issue by precise gene disruption, although the in vivo application remains hindered by the lack of a safe and efficient delivery strategy. Herein, we developed a cell membrane biomimetic core-shell system for light-controllable, precise gene editing. The inner core of the system comprises protamine for CRISPR-Cas9/sgRNA plasmid (pCas9) loading and calcium ions for efficient pCas9 transfection. The shell of the system is camouflaged by a cell membrane and modified with AS1411 aptamers for tumor targeting and photosensitizers to induce lysosomal escape and pCas9 release through reactive oxygen species production, thereby producing light-controllable enhanced gene editing. Neoplastic H1299 cells were reprogrammed using the biomimetic gene editing system upon laser irradiation with reduced VEGF and Vimentin expression, leading to enhanced antimetastatic effects. Genetic disruption of HIF-1α augmented the in vivo chemotherapeutic efficacy of paclitaxel. Our approach of using a membrane-camouflaged system combined with light augmentation provides a potential solution for the in vivo delivery of CRISPR-Cas9 as well as a feasible strategy for cancer therapy.
Collapse
Affiliation(s)
- Lei Qiao
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Min Gao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xiaoqing Yi
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China
| | - Hui Peng
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Ruijie Zhang
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Wanqing Yao
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Xiaoyan He
- School of Life Sciences, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
33
|
Ren XH, Han D, He XY, Guo T, Chen XS, Pang X, Cheng SX. Multi-Targeting Nano-Systems Targeting Heterogeneous Cancer Cells for Therapeutics and Biomarker Detection. Adv Healthc Mater 2023; 12:e2202155. [PMID: 36333906 DOI: 10.1002/adhm.202202155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/15/2022] [Indexed: 11/06/2022]
Abstract
Cancer heterogeneity plays a vital part in cancer resistance and metastasis. To provide a reliable approach to exert a therapy action and evaluate its efficiency in heterogeneous cancer cells, a multiple targeting delivery vector composed of histone encapsulating the therapeutic or diagnostic agent, hyaluronic acid targeting CD44 overexpressed in stem tumor cells, SYL3C aptamer targeting epithelial cell adhesion molecule (EpCAM) overexpressed in epithelial cancer cells, and CL4 aptamer targeting epidermal growth factor receptor (EGFR) overexpressed in mesenchymal cancer cells, is developed. The vector can efficiently target different cancer cells and circulating tumor cells (CTCs) in the peripheral blood of patients for mucin 1 (MUC1) knockout. Furthermore, the multiple targeting vector can be used to co-encapsulate three types of molecular beacons for probing various mRNA biomarkers at single-cell resolution after genome editing. This study provides an efficient approach for exerting therapeutic actions in heterogeneous cancer cells and assessing the therapeutic efficacy by detection of cancer biomarkers via liquid biopsy.
Collapse
Affiliation(s)
- Xiao-He Ren
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, Hubei, 430072, China
| | - Di Han
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, Hubei, 430072, China
| | - Xiao-Yan He
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230011, China
| | - Tao Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, Anhui, 230011, China
| | - Xue-Si Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Xuan Pang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Si-Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, Hubei, 430072, China
| |
Collapse
|
34
|
Pu Y, Wu W, Xiang H, Chen Y, Xu H. CRISPR/Cas9-based genome editing for multimodal synergistic cancer nanotherapy. NANO TODAY 2023; 48:101734. [DOI: 10.1016/j.nantod.2022.101734] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
|
35
|
Gao Y, Wang K, Zhang J, Duan X, Sun Q, Men K. Multifunctional nanoparticle for cancer therapy. MedComm (Beijing) 2023; 4:e187. [PMID: 36654533 PMCID: PMC9834710 DOI: 10.1002/mco2.187] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 01/14/2023] Open
Abstract
Cancer is a complex disease associated with a combination of abnormal physiological process and exhibiting dysfunctions in multiple systems. To provide effective treatment and diagnosis for cancer, current treatment strategies simultaneously focus on various tumor targets. Based on the rapid development of nanotechnology, nanocarriers have been shown to exhibit excellent potential for cancer therapy. Compared with nanoparticles with single functions, multifunctional nanoparticles are believed to be more aggressive and potent in the context of tumor targeting. However, the development of multifunctional nanoparticles is not simply an upgraded version of the original function, but involves a sophisticated system with a proper backbone, optimized modification sites, simple preparation method, and efficient function integration. Despite this, many well-designed multifunctional nanoparticles with promising therapeutic potential have emerged recently. Here, to give a detailed understanding and analyzation of the currently developed multifunctional nanoparticles, their platform structures with organic or inorganic backbones were systemically generalized. We emphasized on the functionalization and modification strategies, which provide additional functions to the nanoparticle. We also discussed the application combination strategies that were involved in the development of nanoformulations with functional crosstalk. This review thus provides an overview of the construction strategies and application advances of multifunctional nanoparticles.
Collapse
Affiliation(s)
- Yan Gao
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Kaiyu Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Xingmei Duan
- Department of PharmacyPersonalized Drug Therapy Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuan ProvinceChina
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| |
Collapse
|
36
|
Laomeephol C, Areecheewakul S, Tawinwung S, Suppipat K, Chunhacha P, Neves NM, Luckanagul JA. Potential roles of hyaluronic acid in in vivo CAR T cell reprogramming for cancer immunotherapy. NANOSCALE 2022; 14:17821-17840. [PMID: 36472072 DOI: 10.1039/d2nr05949e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has recently shown unprecedented clinical efficacy for cancer treatment, particularly of hematological malignancies. However, the complex manufacturing processes that involve ex vivo genetic modification of autologous T cells limits its therapeutic application. CAR T cells generated in vivo provide a valid alternative immunotherapy, "off-the-shelf", for cancer treatment. This approach requires carriers for the delivery of CAR-encoding constructs, which are plasmid DNA or messenger RNA, to T cells for CAR expression to help eradicate the tumor. As such, there are a growing number of studies reporting gene delivery systems for in vivo CAR T cell therapy based on viral vectors and polymeric nanoparticles. Hyaluronic acid (HA) is a natural biopolymer that can serve for gene delivery, because of its inherent properties of cell recognition and internalization, as well as its biodegradability, biocompatibility, and presence of functional groups for the chemical conjugation of targeting ligands. In this review, the potential of HA in the delivery of CAR constructs is discussed on the basis of previous experience of HA-based nanoparticles for gene therapy. Furthermore, current studies on CAR carriers for in vivo-generated CAR T cells are included, giving an idea of a rational design of HA-based systems for the more efficient delivery of CAR to circulating T cells.
Collapse
Affiliation(s)
- Chavee Laomeephol
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Sudartip Areecheewakul
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Supannikar Tawinwung
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Chulalongkorn University Cancer Immunology Excellence Center, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Koramit Suppipat
- Chulalongkorn University Cancer Immunology Excellence Center, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Chulalongkorn University Stem Cell and Cell Therapy Research Center, Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Preedakorn Chunhacha
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nuno M Neves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark - Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Jittima Amie Luckanagul
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
37
|
Intelligent nanotherapeutic strategies for the delivery of CRISPR system. Acta Pharm Sin B 2022. [DOI: 10.1016/j.apsb.2022.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
38
|
Ma H, Cao M. Designed Peptide Assemblies for Efficient Gene Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:13627-13634. [PMID: 36318179 DOI: 10.1021/acs.langmuir.2c02197] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The safe and efficient delivery of nucleic acids including DNA, mRNA, siRNA, and miRNA into targeted cells is critical for gene therapy. Currently, viral gene vectors are very popular, but they have potential toxicity and insecurity. Therefore, the development of nonviral vectors has attracted considerable research attention. Peptide assemblies are superior candidates for being used as gene vectors by having good biocompatibility, versatile molecular design, excellent assembly capacity, ease of modification, and stimuli responsivity. The de novo designed peptides not only can induce efficient condensation of nucleic acids into compacted nanoparticles and protect them from enzymatic digestion but also can effectively overcome biological barriers and improve gene delivery efficiency through targeted delivery, enhanced cellular uptake, improved endolysosomal escape, and nuclear importation. By having these merits, peptidic gene vectors are developing fast, showing outstanding advantages compared to liposome and polymer vectors. This Perspective focuses on peptidic gene delivery systems by emphasizing the molecular design strategies for meeting the criteria of gene condensation, protection from nuclease degradation, cellular uptake, endolysosomal escape, and so on. The new arising research area of peptide-based artificial viruses for gene and ribonucleoprotein delivery has also been reviewed. The challenges and future perspectives are put forward, aiming to provide a conclusive guide for the development of peptidic delivery systems to achieve efficient gene therapy.
Collapse
Affiliation(s)
- Hongchao Ma
- State Key Laboratory of Heavy Oil Processing and Department of Biological and Energy Chemical Engineering, College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| | - Meiwen Cao
- State Key Laboratory of Heavy Oil Processing and Department of Biological and Energy Chemical Engineering, College of Chemistry and Chemical Engineering, China University of Petroleum (East China), Qingdao 266580, China
| |
Collapse
|
39
|
Li JH, Huang LJ, Zhou HL, Shan YM, Chen FM, Lehto VP, Xu WJ, Luo LQ, Yu HJ. Engineered nanomedicines block the PD-1/PD-L1 axis for potentiated cancer immunotherapy. Acta Pharmacol Sin 2022; 43:2749-2758. [PMID: 35484402 PMCID: PMC9622913 DOI: 10.1038/s41401-022-00910-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022]
Abstract
Immunotherapy, in particular immune checkpoint blockade (ICB) therapy targeting the programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) axis, has remarkably revolutionized cancer treatment in the clinic. Anti-PD-1/PD-L1 therapy is designed to restore the antitumor response of cytotoxic T cells (CTLs) by blocking the interaction between PD-L1 on tumour cells and PD-1 on CTLs. Nevertheless, current anti-PD-1/PD-L1 therapy suffers from poor therapeutic outcomes in a large variety of solid tumours due to insufficient tumour specificity, severe cytotoxic effects, and the occurrence of immune resistance. In recent years, nanosized drug delivery systems (NDDSs), endowed with highly efficient tumour targeting and versatility for combination therapy, have paved a new avenue for cancer immunotherapy. In this review article, we summarized the recent advances in NDDSs for anti-PD-1/PD-L1 therapy. We then discussed the challenges and further provided perspectives to promote the clinical application of NDDS-based anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Jun-Hao Li
- College of Sciences, Shanghai University, Shanghai, 200444, China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lu-Jia Huang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Hui-Ling Zhou
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi-Ming Shan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fang-Min Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Vesa-Pekka Lehto
- Department of Applied Physics, University of Eastern Finland, 70211, Kuopio, Finland
| | - Wu-Jun Xu
- Department of Applied Physics, University of Eastern Finland, 70211, Kuopio, Finland.
| | - Li-Qiang Luo
- College of Sciences, Shanghai University, Shanghai, 200444, China
| | - Hai-Jun Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
40
|
Zhou L, Liang H, Ge Y, Ding W, Chen Q, Zhang T, Xiao L, Li Y, Dong J, He X, Xue F, Jiang L. Precisely Targeted Nano-Controller of PD-L1 Level for Non-Small Cell Lung Cancer Spinal Metastasis Immunotherapy. Adv Healthc Mater 2022; 11:e2200938. [PMID: 35904523 DOI: 10.1002/adhm.202200938] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/07/2022] [Indexed: 01/28/2023]
Abstract
Although immune checkpoint inhibitors (ICIs) have been widely applied to treat non-small cell lung cancer (NSCLC), a significant proportion of patients, especially those with spinal metastasis (NSCLC-SM), are insensitive to anti-programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) ICIs. A drug delivery nano-controller of PD-L1 that targets NSCLC-SM can solve this problem, however, none have been developed to date. In this study, it is shown that integrin β3 (β3-int) is strongly upregulated in NSCLC-SM. Its inhibitor RGDyK promotes PD-L1 ubiquitination, indicating the potential application of RGDyK as a new PD-L1 inhibitor in nano-controller and a targeting peptide for NSCLC-SM treatment. According to the synergistic effect of photodynamic therapy and ICIs on T-cell activation through the release of tumor antigens, RGDyK-modified and zinc protoporphyrin (ZnPP)-loaded mesoporous silicon nanoparticles (ZnPP@MSN-RGDyK) are fabricated. The ZnPP@MSN-RGDyK nanoparticles precisely target β3-int to inhibit PD-L1, exhibiting high photodynamic therapy efficiency, and excellent immunotherapeutic effects in an NSCLC-SM mouse model. Collectively, the findings indicate that ZnPP@MSN-RGDyK is a promising immunotherapeutic agent for treating NSCLC-SM.
Collapse
Affiliation(s)
- Lei Zhou
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.,Cancer center, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Haifeng Liang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.,Cancer center, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Yuxiang Ge
- Department of Orthopaedics Surgery, Minhang Hospital, School of Materials Science and Engineering, Fudan University, Shanghai, 200237, P. R. China
| | - Wang Ding
- Department of Orthopaedics Surgery, Minhang Hospital, School of Materials Science and Engineering, Fudan University, Shanghai, 200237, P. R. China
| | - Qing Chen
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.,Cancer center, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, P. R. China
| | - Taiwei Zhang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.,Cancer center, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, P. R. China
| | - Lan Xiao
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, 4059, Australia
| | - Yulin Li
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomedical Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Jian Dong
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.,Cancer center, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.,Department of Orthopaedic Surgery, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, 200940, P. R. China
| | - Xiaowen He
- Department of Orthopaedic Surgery, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, 200940, P. R. China
| | - Fengfeng Xue
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, P. R. China
| | - Libo Jiang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.,Cancer center, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| |
Collapse
|
41
|
Kim J, Thomas SN. Opportunities for Nitric Oxide in Potentiating Cancer Immunotherapy. Pharmacol Rev 2022; 74:1146-1175. [PMID: 36180108 PMCID: PMC9553106 DOI: 10.1124/pharmrev.121.000500] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 05/15/2022] [Accepted: 07/05/2022] [Indexed: 11/22/2022] Open
Abstract
Despite nearly 30 years of development and recent highlights of nitric oxide (NO) donors and NO delivery systems in anticancer therapy, the limited understanding of exogenous NO's effects on the immune system has prevented their advancement into clinical use. In particular, the effects of exogenously delivered NO differing from that of endogenous NO has obscured how the potential and functions of NO in anticancer therapy may be estimated and exploited despite the accumulating evidence of NO's cancer therapy-potentiating effects on the immune system. After introducing their fundamentals and characteristics, this review discusses the current mechanistic understanding of NO donors and delivery systems in modulating the immunogenicity of cancer cells as well as the differentiation and functions of innate and adaptive immune cells. Lastly, the potential for the complex modulatory effects of NO with the immune system to be leveraged for therapeutic applications is discussed in the context of recent advancements in the implementation of NO delivery systems for anticancer immunotherapy applications. SIGNIFICANCE STATEMENT: Despite a 30-year history and recent highlights of nitric oxide (NO) donors and delivery systems as anticancer therapeutics, their clinical translation has been limited. Increasing evidence of the complex interactions between NO and the immune system has revealed both the potential and hurdles in their clinical translation. This review summarizes the effects of exogenous NO on cancer and immune cells in vitro and elaborates these effects in the context of recent reports exploiting NO delivery systems in vivo in cancer therapy applications.
Collapse
Affiliation(s)
- Jihoon Kim
- Parker H. Petit Institute for Bioengineering and Bioscience (J.K., S.N.T.), George W. Woodruff School of Mechanical Engineering (J.K., S.N.T.), and Wallace H. Coulter Department of Biomedical Engineering (S.N.T.), Georgia Institute of Technology, Atlanta, Georgia; Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia (S.N.T.); and Division of Biological Science and Technology, Yonsei University, Wonju, South Korea (J.K.)
| | - Susan N Thomas
- Parker H. Petit Institute for Bioengineering and Bioscience (J.K., S.N.T.), George W. Woodruff School of Mechanical Engineering (J.K., S.N.T.), and Wallace H. Coulter Department of Biomedical Engineering (S.N.T.), Georgia Institute of Technology, Atlanta, Georgia; Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia (S.N.T.); and Division of Biological Science and Technology, Yonsei University, Wonju, South Korea (J.K.)
| |
Collapse
|
42
|
Yin X, Cheng Y, Feng Y, Stiles WR, Park SH, Kang H, Choi HS. Phototheranostics for multifunctional treatment of cancer with fluorescence imaging. Adv Drug Deliv Rev 2022; 189:114483. [PMID: 35944585 PMCID: PMC9860309 DOI: 10.1016/j.addr.2022.114483] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/06/2022] [Accepted: 08/03/2022] [Indexed: 01/25/2023]
Abstract
Phototheranostics stem from the recent advances in nanomedicines and bioimaging to diagnose and treat human diseases. Since tumors' diversity, heterogeneity, and instability limit the clinical application of traditional diagnostics and therapeutics, phototheranostics, which combine light-induced therapeutic and diagnostic modalities in a single platform, have been widely investigated. Numerous efforts have been made to develop phototheranostics for efficient light-induced antitumor therapeutics with minimal side effects. Herein, we review the fundamentals of phototheranostic nanomedicines with their biomedical applications. Furthermore, the progress of near-infrared fluorescence imaging and cancer treatments, including photodynamic therapy and photothermal therapy, along with chemotherapy, immunotherapy, and gene therapy, are summarized. This review also discusses the opportunities and challenges associated with the clinical translation of phototheranostics in pan-cancer research. Phototheranostics can pave the way for future research, improve the quality of life, and prolong cancer patients' survival times.
Collapse
Affiliation(s)
- Xiaoran Yin
- Department of Oncology, The Second Affiliate Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China,Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Yifan Cheng
- Department of Oncology, The Second Affiliate Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Yan Feng
- Department of Oncology, The Second Affiliate Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Wesley R. Stiles
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Seung Hun Park
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Homan Kang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA,Corresponding authors at: 149 13th Street, Boston, MA 02129, USA., (H. Kang), (H.S. Choi)
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA,Corresponding authors at: 149 13th Street, Boston, MA 02129, USA., (H. Kang), (H.S. Choi)
| |
Collapse
|
43
|
Khademi Z, Ramezani M, Alibolandi M, Zirak MR, Salmasi Z, Abnous K, Taghdisi SM. A novel dual-targeting delivery system for specific delivery of CRISPR/Cas9 using hyaluronic acid, chitosan and AS1411. Carbohydr Polym 2022; 292:119691. [PMID: 35725215 DOI: 10.1016/j.carbpol.2022.119691] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/13/2022] [Accepted: 05/31/2022] [Indexed: 11/18/2022]
Abstract
A facile method was designed that can specifically deliver CRISPR/Cas9 into target cells nuclei and reduce the off-target effects. A multifunctional delivery vector for FOXM1 knockout was composed by integration of cell targeting polymer (hyaluronic acid) and cell and nuclear targeting group (AS1411 aptamer) on the surface of nanoparticles formed by genome editing plasmid and chitosan (CS) as the core (Apt-HA-CS-CRISPR/Cas9). The data of cytotoxicity experiment and western blot confirmed this issue. The results of flow cytometry analysis and fluorescence imaging demonstrated that Apt-HA-CS-CRISPR/Cas9 was significantly internalized into target cells (MCF-7, SK-MES-1, HeLa) but not into nontarget cells (HEK293). Furthermore, the in vivo studies displayed that the Apt-HA-CS-CRISPR/Cas9 was strongly rendered tumor inhibitory effect and delivered efficiently CRISPR/Cas9 into the tumor with no detectable distribution in other organs compared with naked plasmid. This approach provides an avenue for specific in vivo gene editing therapeutics with the lowest side effect.
Collapse
Affiliation(s)
- Zahra Khademi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Zirak
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Salmasi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
44
|
Caprifico AE, Foot PJS, Polycarpou E, Calabrese G. Advances in Chitosan-Based CRISPR/Cas9 Delivery Systems. Pharmaceutics 2022; 14:pharmaceutics14091840. [PMID: 36145588 PMCID: PMC9505239 DOI: 10.3390/pharmaceutics14091840] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/02/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Clustered regularly interspaced short palindromic repeat (CRISPR) and the associated Cas endonuclease (Cas9) is a cutting-edge genome-editing technology that specifically targets DNA sequences by using short RNA molecules, helping the endonuclease Cas9 in the repairing of genes responsible for genetic diseases. However, the main issue regarding the application of this technique is the development of an efficient CRISPR/Cas9 delivery system. The consensus relies on the use of non-viral delivery systems represented by nanoparticles (NPs). Chitosan is a safe biopolymer widely used in the generation of NPs for several biomedical applications, especially gene delivery. Indeed, it shows several advantages in the context of gene delivery systems, for instance, the presence of positively charged amino groups on its backbone can establish electrostatic interactions with the negatively charged nucleic acid forming stable nanocomplexes. However, its main limitations include poor solubility in physiological pH and limited buffering ability, which can be overcome by functionalising its chemical structure. This review offers a critical analysis of the different approaches for the generation of chitosan-based CRISPR/Cas9 delivery systems and suggestions for future developments.
Collapse
|
45
|
Peng H, Qiao L, Shan G, Gao M, Zhang R, Yi X, He X. Stepwise responsive carboxymethyl chitosan-based nanoplatform for effective drug-resistant breast cancer suppression. Carbohydr Polym 2022; 291:119554. [DOI: 10.1016/j.carbpol.2022.119554] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 04/18/2022] [Accepted: 04/27/2022] [Indexed: 12/13/2022]
|
46
|
Wang ZY, Li DL, Tian X, Li Y, Zhang CY. Single-Molecule Counting of FTO in Human Breast Tissues Based on a Rolling Circle Transcription Amplification-Driven Clustered Regularly Interspaced Short Palindromic Repeat─Cas12a. Anal Chem 2022; 94:11425-11432. [PMID: 35916620 DOI: 10.1021/acs.analchem.2c02578] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
N6-methyladenosine modification as an mRNA modification in mammalian cells is dynamically reversible, regulated by RNA demethylase [e.g., fat mass and obesity-associated protein (FTO)]. The abnormal expression of FTO is closely related to numerous diseases (e.g., various cancers and obesity). Herein, we demonstrate the single-molecule counting of FTO in human cancer cells and breast tissues based on a T7 RNA polymerase-mediated rolling circle transcription (RCT) amplification-driven clustered regularly interspaced short palindromic repeat (CRISPR)─Cas12a. When FTO is present, it demethylates the DNA substrate, initiating the DpnII-mediated cleavage reaction. After magnetic separation, the cleaved DNA fragments trigger the T7 RNA polymerase-mediated RCT amplification, activating CRISPR-/Cas12a-mediated cleavage of signal probes and releasing abundant FAM molecules that are simply counted via single-molecule detection. In this assay, only target FTO can generate CRISPR RNAs, efficiently improving detection specificity. Moreover, the integration of single-molecule detection with magnetic separation achieves zero background and effectively enhances detection sensitivity. This method can specifically and sensitively monitor FTO activity with a limit of detection of 1.20 × 10-13 M, and it may measure FTO at the single-cell level. Furthermore, it may accurately discriminate the FTO expression level in breast tissues between healthy persons and breast cancer patients and screen the FTO inhibitors as well, with great potential in clinical diagnosis and drug discovery.
Collapse
Affiliation(s)
- Zi-Yue Wang
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| | - Dong-Ling Li
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| | - Xiaorui Tian
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| | - Yueying Li
- Institute of Immunity and Infectious Diseases, School of Medicine and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Chun-Yang Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
47
|
Hejabi F, Abbaszadeh MS, Taji S, O’Neill A, Farjadian F, Doroudian M. Nanocarriers: A novel strategy for the delivery of CRISPR/Cas systems. Front Chem 2022; 10:957572. [PMID: 36092658 PMCID: PMC9450496 DOI: 10.3389/fchem.2022.957572] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 12/14/2022] Open
Abstract
In recent decades, clustered regularly interspaced short palindromic repeat/CRISPR-associated protein (CRISPR/Cas) has become one of the most promising genome-editing tools for therapeutic purposes in biomedical and medical applications. Although the CRISPR/Cas system has truly revolutionized the era of genome editing, the safe and effective delivery of CRISPR/Cas systems represents a substantial challenge that must be tackled to enable the next generation of genetic therapies. In addition, there are some challenges in the in vivo delivery to the targeted cells/tissues. Nanotechnology-based drug delivery systems can be employed to overcome this issue. This review discusses different types and forms of CRISPR/Cas systems and the current CRISPR/Cas delivery systems, including non-viral carriers such as liposomes, polymeric, and gold particles. The focus then turns to the viral nanocarriers which have been recently used as a nanocarrier for CRISPR/Cas delivery.
Collapse
Affiliation(s)
- Faranak Hejabi
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | | | - Shirinsadat Taji
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Andrew O’Neill
- Department of Clinical Medicine, Tallaght University Hospital and Trinity College Dublin, Dublin, Ireland
| | - Fatemeh Farjadian
- Pharmaceutical Sciences Research Center, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Doroudian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
- *Correspondence: Mohammad Doroudian,
| |
Collapse
|
48
|
Shalaby KE, Aouida M, Gupta V, Abdesselem H, El-Agnaf OMA. Development of non-viral vectors for neuronal-targeted delivery of CRISPR-Cas9 RNA-proteins as a therapeutic strategy for neurological disorders. Biomater Sci 2022; 10:4959-4977. [PMID: 35880637 DOI: 10.1039/d2bm00368f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The aging population contributes to an increase in the prevalence of neurodegenerative diseases, such as Parkinson's disease (PD). Due to the progressive nature of these diseases and an incomplete understanding of their pathophysiology, current drugs are inefficient, with a limited efficacy and major side effects. In this study, CRISPR-Cas9 RNA-proteins (RNP) composed of a Cas9 nuclease and single-guide RNA were delivered with a non-viral targeted delivery system to rescue the PD-associated phenotype in neuronal cells. Here, we fused the cell-penetrating amphipathic peptide, PepFect14 (PF14), with a short fragment of the rabies virus glycoprotein (C2) previously shown to have an affinity towards nicotinic acetylcholine receptors expressed on neuronal cells and on the blood-brain barrier. The resultant peptide, C2-PF14, was used to complex with and deliver RNPs to neuronal cells. We observed that RNP/C2-PF14 complexes formed nanosized, monodispersed, and nontoxic nanoparticles that led to a specific delivery into neuronal cells. α-Synuclein (α-syn) plays a major role in the pathology of PD and is considered to be a target for therapy. We demonstrated that CRISPR/Cas9 RNP delivered by C2-PF14 achieved α-syn gene (SNCA) editing in neuronal cells as determined by T7EI assay and western blotting. Furthermore, RNP/C2-PF14 relieved PD-associated toxicity in neuronal cells in vitro. This is a proof-of-concept towards simple and safe targeted genome-editing for treating PD and other neurological disorders.
Collapse
Affiliation(s)
- Karim E Shalaby
- Biological and Biomedical Sciences Division, College of Health & Life Sciences, Hamad Bin Khalifa University, Doha, Qatar. .,Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Mustapha Aouida
- Biological and Biomedical Sciences Division, College of Health & Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| | - Vijay Gupta
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Houari Abdesselem
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Omar M A El-Agnaf
- Biological and Biomedical Sciences Division, College of Health & Life Sciences, Hamad Bin Khalifa University, Doha, Qatar. .,Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| |
Collapse
|
49
|
Chen QB, Shen MH, Ren XH, Zhu S, Shang JT, Liu W, Zhang ZW, Dong ZJ, Gu HZ, Zhang XZ, Yuan Q, Zou T. Tumor-triggered targeting ammonium bicarbonate liposomes for tumor multimodal therapy. J Mater Chem B 2022; 10:5154-5164. [PMID: 35666635 DOI: 10.1039/d2tb00409g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tumor-triggered targeting ammonium bicarbonate (TTABC) liposomes were proposed to improve the uptake of ammonium bicarbonate (ABC) liposomes in tumor cells and retain their long circulation in vivo in our previous study. However, it must be solved how to precisely release the loaded drugs of the TTABC liposomes into tumor cells. In addition, synergistic multimodal therapy could result in better tumor treatment outcomes than monomodal chemotherapy. In the research, we prepared indocyanine green (ICG) and doxorubicin (DOX) encapsulated TTABC liposomes (ICG&DOX@TTABC) to achieve near-infrared (NIR) light-controlled chemo/photothermal/photodynamic multimodal therapy guided by fluorescence and photothermal imaging. In vitro and vivo studies show that ICG&DOX@TTABC can specifically accumulate in tumor tissues, effectively transform NIR light into local thermo-therapy, and have excellent anti-tumor ability without obvious side effects. ICG&DOX@TTABC could be promising for fluorescence and photothermal imaging-guided chemo/photothermal/photodynamic tumor treatment.
Collapse
Affiliation(s)
- Quan-Bing Chen
- State Key Laboratory of Refractories and Metallurgy, Key Laboratory of Coal Conversion & New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China.
| | - Mei-Hua Shen
- Institute of Pharmaceutical Innovation, College of Medicine, Wuhan University of Science and Technology, Wuhan 430065, P. R. China.
| | - Xiao-He Ren
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Shuai Zhu
- State Key Laboratory of Refractories and Metallurgy, Key Laboratory of Coal Conversion & New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China.
| | - Jin-Ting Shang
- School of Medicine, Jianghan University, Wuhan 430056, P. R. China
| | - Wei Liu
- School of Medicine, Jianghan University, Wuhan 430056, P. R. China
| | - Zhao-Wei Zhang
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Key Laboratory of Detection for Mycotoxins, Ministry of Agriculture and Rural Affairs, National Reference Laboratory for Agricultural Testing (Biotoxin), Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan 430062, P. R. China
| | - Zhi-Jun Dong
- State Key Laboratory of Refractories and Metallurgy, Key Laboratory of Coal Conversion & New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China.
| | - Hua-Zhi Gu
- State Key Laboratory of Refractories and Metallurgy, Key Laboratory of Coal Conversion & New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China.
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Qiong Yuan
- Institute of Pharmaceutical Innovation, College of Medicine, Wuhan University of Science and Technology, Wuhan 430065, P. R. China.
| | - Tao Zou
- State Key Laboratory of Refractories and Metallurgy, Key Laboratory of Coal Conversion & New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, P. R. China.
| |
Collapse
|
50
|
Ren X, He X, Xu C, Han D, Cheng S. Functional Tumor Targeting Nano-Systems for Reprogramming Circulating Tumor Cells with In Situ Evaluation on Therapeutic Efficiency at the Single-Cell Level. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105806. [PMID: 35595716 PMCID: PMC9313495 DOI: 10.1002/advs.202105806] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/20/2022] [Indexed: 05/03/2023]
Abstract
Tumor heterogeneity is primarily responsible for treatment resistance and cancer relapses. Being critically important to address this issue, the timely evaluation of the appropriateness of therapeutic actions at the single-cell level is still facing challenges. By using multi-functionalized nano-systems with the delivery vector composed of histone for plasmids loading, hyaluronic acid for tumor targeting, and a fusion peptide for C-X-C motif chemokine receptor 4 (CXCR4) targeting as well as nuclear localization, the reprogramming of circulating tumor cells (CTCs) with in situ detection on biomarkers at the single-cell level is realized. By efficient co-delivery of the genome editing plasmid for CXCR4 knockout and molecular beacons for detection of upregulated mRNA biomarkers into CTCs in unprocessed whole blood, the therapeutic outcomes of genome editing at the single-cell level can be in situ evaluated. The single-cell analysis shows that CXCR4 in CTCs of cancer patients is efficiently downregulated, resulting in upregulated anticancer biomarkers such as p53 and p21. The study provides a facile strategy for in-depth profiling of cancer cell responses to therapeutic actions at single-cell resolution to evaluate the outcomes of treatments timely and conveniently.
Collapse
Affiliation(s)
- Xiao‐He Ren
- Key Laboratory of Biomedical Polymers of Ministry of EducationDepartment of ChemistryWuhan UniversityWuhan430072P. R. China
| | - Xiao‐Yan He
- School of Life SciencesAnhui Medical UniversityHefei230032P. R. China
| | - Chang Xu
- Key Laboratory of Biomedical Polymers of Ministry of EducationDepartment of ChemistryWuhan UniversityWuhan430072P. R. China
| | - Di Han
- Key Laboratory of Biomedical Polymers of Ministry of EducationDepartment of ChemistryWuhan UniversityWuhan430072P. R. China
| | - Si‐Xue Cheng
- Key Laboratory of Biomedical Polymers of Ministry of EducationDepartment of ChemistryWuhan UniversityWuhan430072P. R. China
| |
Collapse
|