1
|
Baldan M, Zhang S, Sun Q, Su Y, Mei D, Sun R, Zheng A, Liu D, Zhang J, Huo R, Tian Y, Han L, Wang S, Wang Y, Cui C. NIR-Triggered siRNA Release and Lysosomal Escape for Synergistic Photothermal Tumor Therapy. Int J Nanomedicine 2025; 20:4863-4882. [PMID: 40259913 PMCID: PMC12010081 DOI: 10.2147/ijn.s511655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/26/2025] [Indexed: 04/23/2025] Open
Abstract
Background Resistance to traditional treatments has spurred research into innovative therapeutic approaches for tumors. Among these innovative treatments, photothermal therapy (PTT) has gained increasing attention for its use of photothermal agents (PTAs) to convert light into heat for localized tumor ablation. However, PTT faces limitations due to heat shock protein 70 (HSP70)-mediated resistance in tumor cells. Combining PTT via indocyanine green (ICG) with siRNA HSP70 could reduce the thermal resistance of the tumor, thereby enhancing treatment efficacy. Albumin-based nanoparticles (NPs) can effectively deliver ICG and siRNA into tumor cells. When exposed to near-infrared (NIR) light, these nanoparticles trigger lysosomal escape and release, further enhancing gene silencing activity. Methods This study aimed to develop a biocompatible delivery system, HSA@ICG/siRNA NPs, for photothermal-enhanced tumor therapy. The nanoparticles were characterized for size, charge, surface functionalization, and photoconversion properties. In vitro antitumor efficacy was evaluated using MTT assay, calcein AM/PI staining, RT-PCR, and Western blot in 4T1 tumor cells. In vivo, we assessed photothermal effects, biodistribution, tumor inhibition, and biosafety following irradiation. Results Characterization confirmed the successful synthesis of uniform, stable HSA@ICG/siRNA NPs with effective photothermal conversion properties. Cellular uptake studies revealed high siRNA internalization, with laser-induced lysosomal escape enhancing cytoplasmic delivery. In vitro, gene silencing reduced mRNA and protein levels by 82.8% and 65%, respectively. In vivo, local tumor temperature increased to 42°C within 3 minutes, indicating a mild but effective photothermal effect. Tumor inhibition rates were 50.00% ± 9.16% for HSA@ICG and 71.26% ± 7.92% for HSA@ICG/siRNA, demonstrating enhanced tumor suppression. The treatment achieved sustained tumor targeting with minimal off-target toxicity. Conclusion As a dual-function photothermal therapy agent, HSA@ICG/siRNA NPs combine targeted gene silencing with photothermal effects, demonstrating significant therapeutic promise. This integrated approach addresses tumor resistance, offering a potential advancement in cancer treatment strategies.
Collapse
Affiliation(s)
- Myagmarsuren Baldan
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Shuang Zhang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Qi Sun
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Yan Su
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
- Medical Oncology Department, Pediatric Oncology Center, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| | - Dong Mei
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
- Department of Pharmacy, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, People’s Republic of China
| | - Ran Sun
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Ao Zheng
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Danni Liu
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Jie Zhang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Ran Huo
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Yang Tian
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Le Han
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Shibo Wang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Yaoqi Wang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Chunying Cui
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, People’s Republic of China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
2
|
Razzaq S, Fatima I, Moafian Z, Rahdar A, Fathi-Karkan S, Kharaba Z, Shirzad M, Khan A, Pandey S. Nanomedicine innovations in colon and rectal cancer: advances in targeted drug and gene delivery systems. Med Oncol 2025; 42:113. [PMID: 40097759 DOI: 10.1007/s12032-025-02670-z] [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: 01/01/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Nanotechnology has revolutionized cancer diagnostics and therapy, offering unprecedented possibilities to overcome the constraints of conventional treatments. This study provides a detailed overview of the current progress and difficulties in the creation of nanostructured materials, with a specific emphasis on their use in drug and gene delivery systems. The study examines tactics that attempt to improve the effectiveness and safety of chemotherapeutic drugs such as doxorubicin (Dox) by focusing on the potential of antibody-drug conjugates and functionalized nanoparticles. Moreover, it clarifies the challenges encountered in administering nanoparticles orally for gastrointestinal treatments, emphasizing the crucial physicochemical properties that affect their behavior in the gastrointestinal system. This study highlights the transformational potential of nanostructured materials in precision oncology by examining advanced breakthroughs such cell membrane-camouflaged nanoparticles and inorganic nanoparticles designed for gastrointestinal disorders. The text investigates the processes involved in the absorption of nanoparticles and their destruction in lysosomes, revealing the many methods in which enterocytes take up these particles. This study strongly supports the use of advanced nanoparticle-based methods to reduce the harmful effects on the whole body and improve the effectiveness of therapy, based on a thorough examination of current experiments on animals and humans. The main objective of this paper is to provide a fundamental comprehension that will stimulate more investigation and practical use in the field of cancer nanomedicine, advancing its boundaries.
Collapse
Affiliation(s)
- Sobia Razzaq
- School of Pharmacy, University of Management and Technology, Lahore, Punjab, Pakistan
| | - Iqra Fatima
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Zeinab Moafian
- Department of Chemistry and Biochemistry, University of Delaware, Newark, USA
| | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol, 538-98615, Iran.
| | - Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran.
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 9414974877, Iran.
| | - Zelal Kharaba
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, UAE
| | - Maryam Shirzad
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ahmad Khan
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| | - Sadanand Pandey
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, 173229, Himachal Pradesh, India.
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk, 38541, Republic of Korea.
| |
Collapse
|
3
|
Liu J, Xiong L, Hu Y, Wang Z, Dai J, Li T, Guo X, Liu R, Yu Z, Li Y, Li Y. Probing Structural Variants of Irregular DNA G-Tracts ( N ≤ 2) Using MspA Nanopores. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 39977584 DOI: 10.1021/acsami.4c19806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Guanine-rich DNA sequences with short G-tracts (n ≤ 2) are highly prevalent and abundant in the human genome, some of which are found to be associated with diseases (Maity et al. Nucleic Acids Res. 2020, 48 (6), 3315-3327). Unlike conventional G-quadruplexes with three or more folded layers, these sequences with G2 tracts featuring two bilayered blocks remain largely unexplored. Here, we employed nanopore experiments and all-atom molecular dynamics simulations to investigate the unwinding strengths and dynamics of these bilayered blocks. Our results demonstrated that in an electric field, the tumor-targeting element AS1411, along with its derivatives AT11 and Z-G4, strongly interacted with the M2-MspA nanopore, resulting in at least two distinct populations (types I and II events) characterized by different current blockage fractions and dwell times. Despite AS1411 being well characterized with up to eight secondary structures by nuclear magnetic resonance spectroscopy, our nanopore experiments revealed only two populations. This could be reasonably explained by (i) reversible docking with high rigidity and (ii) strand separation and translocation. Notably, a new event type (type III) for Z-G4 suggested reduced susceptibility in the last layer, contributing to its increased rigidity. Furthermore, voltage-dependent dynamics revealed that Z-G4 exhibited extended dwell times for docking and partial unwinding, unlike AT11. Our in-solution nanopore experiments and MD simulation results would benefit toward understanding the folding principles of complicated structural variants by sequences consisting of multiple short G-tracts, paving the way for the rapid identification of similar-sequence nucleic acid aptamers in molecular diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Jiadun Liu
- School of Microelectronics, MOE Engineering Research Center of Integrated Circuits for Next Generation Communications, Southern University of Science and Technology, Shenzhen 518055, China
| | - Luoan Xiong
- School of Physics and Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, and Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300071, China
| | - Yuhang Hu
- School of Microelectronics, MOE Engineering Research Center of Integrated Circuits for Next Generation Communications, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhuofei Wang
- School of Microelectronics, MOE Engineering Research Center of Integrated Circuits for Next Generation Communications, Southern University of Science and Technology, Shenzhen 518055, China
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Jing Dai
- School of Microelectronics, MOE Engineering Research Center of Integrated Circuits for Next Generation Communications, Southern University of Science and Technology, Shenzhen 518055, China
- School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Tie Li
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xinrong Guo
- School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Ronghui Liu
- School of Microelectronics, MOE Engineering Research Center of Integrated Circuits for Next Generation Communications, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhongbo Yu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Yao Li
- School of Physics and Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, and Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300071, China
| | - Yi Li
- School of Microelectronics, MOE Engineering Research Center of Integrated Circuits for Next Generation Communications, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
4
|
Shokoohi M, Sedaghatshoar S, Arian H, Mokarami M, Habibi F, Bamarinejad F. Genetic advancements in breast cancer treatment: a review. Discov Oncol 2025; 16:127. [PMID: 39918655 PMCID: PMC11805739 DOI: 10.1007/s12672-025-01884-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025] Open
Abstract
Breast cancer (BC) remains a leading cause of cancer-related deaths among women globally, highlighting the urgent need for more effective and targeted therapies. Traditional treatments, including surgery, chemotherapy, and radiation, face limitations such as drug resistance, metastasis, and severe side effects. Recent advancements in gene therapy, particularly CRISPR/Cas9 technology and Oncolytic Virotherapy (OVT), are transforming the BC treatment landscape. CRISPR/Cas9 enables precise gene editing to correct mutations in oncogenes like HER2 and MYC, directly addressing tumor growth and immune evasion. Simultaneously, OVT leverages genetically engineered viruses to selectively destroy cancer cells and stimulate robust antitumor immune responses. Despite their potential, gene therapies face challenges, including off-target effects, delivery issues, and ethical concerns. Innovations in delivery systems, combination strategies, and integrating gene therapy with existing treatments offer promising solutions to overcome these barriers. Personalized medicine, guided by genomic profiling, further enhances treatment precision by identifying patient-specific mutations, such as BRCA1 and BRCA2, allowing for more tailored and effective interventions. As research progresses, the constructive interaction between gene therapy, immunotherapy, and traditional approaches is paving the way for groundbreaking advancements in BC care. Continued collaboration between researchers and clinicians is essential to translate these innovations into clinical practice, ultimately improving BC patients' survival rates and quality of life.
Collapse
Affiliation(s)
- Marzieh Shokoohi
- Department of Life Sciences Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, Iran.
- Amino Techno Gene Virtual Private Laboratory, Tehran, Iran.
| | - Sadaf Sedaghatshoar
- Kent School of Social Work and Family Science, University of Louisville, Louisville, KY, USA
| | - Homaira Arian
- Pharmaceutical Biotechnology Department, Pharmacy Faculty, Anadolu University, Eskishehir, Turkey.
| | - Milad Mokarami
- Student Research Committee, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Fatemeh Habibi
- Department of Speech Therapy, School of Rehabilitation, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bamarinejad
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
5
|
Gharatape A, Amanzadi B, Mohamadi F, Rafieian M, Faridi-Majidi R. Recent advances in polymeric and lipid stimuli-responsive nanocarriers for cell-based cancer immunotherapy. Nanomedicine (Lond) 2024; 19:2655-2678. [PMID: 39540464 DOI: 10.1080/17435889.2024.2416377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Conventional cancer therapy has major limitations, including non-specificity, unavoidable side effects, low specific tumor accumulation and systemic toxicity. In recent years, more effective and precise treatment methods have been developed, including cell-based immunotherapy. Carriers that can accurately and specifically target cells and equip them to combat cancer cells are particularly important for developing this therapy. As a result, attention has been drawn to smart nanocarriers that can react to specific stimuli. Thus, stimuli-responsive nanocarriers have attracted increasing attention because they can change their physicochemical properties in response to stimulus conditions, such as pH, enzymes, redox agents, hypoxia, light and temperature. This review highlights recent advances in various stimuli-responsive nanocarriers, discussing loading, targeted delivery, cellular uptake, biocompatibility and immunomodulation in cell-based immunotherapy. Finally, future challenges and perspectives regarding the possible clinical translation of nanocarriers are discussed.
Collapse
Affiliation(s)
- Alireza Gharatape
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Bentolhoda Amanzadi
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Faranak Mohamadi
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Mahdieh Rafieian
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
| | - Reza Faridi-Majidi
- Advanced Laboratory of Nanocarriers Synthesis, Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1417755469, Iran
- Pharmaceutical Nanotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Jin X, Lopes D, Lopes J, Hua Z, Lei Y, Ghanbari R, Nazarzadeh Zare E, Borzacchiello A, Karimi Male H, Iravani S, Sillanpää M, Prakash C, Wang X, Cláudia Paiva-Santos A, Makvandi P, Xu Y. Bioengineered Abiotic Nanomaterials Through Cell Membrane-Camouflaging: Advancements and Challenges in Lung Cancer. Adv Healthc Mater 2024; 13:e2401525. [PMID: 38978444 DOI: 10.1002/adhm.202401525] [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: 04/25/2024] [Revised: 05/28/2024] [Indexed: 07/10/2024]
Abstract
Lung cancer remains a major global health concern with high mortality rates and poor prognosis. Bridging the gap between the chemical and cellular understanding of cell-decorated biomimetic nanocomposites and their clinical translation is crucial for developing effective therapies. Nanocomposites show promise in targeted drug delivery and diagnostics, but their clinical application is hindered by biocompatibility and clearance issues. To overcome these challenges, biomimetic approaches utilizing cell membrane-coated nanomaterials emerge. By camouflaging nanomaterials with cell membranes, the biointerfaces are enhanced, and the inherent properties of the donor cell membranes are acquired. This review provides an overview of recent advancements on cell membrane-coated nanocomposites for lung cancer diagnosis and treatment. It discusses fabrication techniques, biomedical applications, challenges, and future prospects. The incorporation of cell membranes into nanocomposites holds potential for improved lung cancer therapy, but further development and refinement are needed for precise tumor targeting. Addressing the identified challenges will pave the way for clinical translation of these biomimetic nanoplatforms and advance lung cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Xuru Jin
- Department of Respiratory and Critical Care Medicine, NanoBioMed Group, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Daniela Lopes
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, 3000-548, Portugal
| | - Joana Lopes
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, 3000-548, Portugal
| | - Zhidan Hua
- Department of Respiratory and Critical Care Medicine, NanoBioMed Group, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| | - Ying Lei
- Department of Respiratory and Critical Care Medicine, NanoBioMed Group, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| | - Roham Ghanbari
- School of Chemistry, College of Science, University of Tehran, Tehran, 14174-66191, Iran
| | | | - Assunta Borzacchiello
- Institute of Polymers, Composites and Biomaterials, National Research Council (IPCB-CNR), Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Hassan Karimi Male
- School of Resources and Environment, University of Electronic Science and Technology of China, P.O. Box 611731, Xiyuan Ave, Chengdu, 610054, P. R. China
- Department of chemical engineering, Quchan university of Technology, Quchan, Iran
| | - Siavash Iravani
- Independent Researcher, W Nazar ST, Boostan Ave, Isfahan, Iran
| | - Mika Sillanpää
- Functional Materials Group, Gulf University for Science and Technology, Mubarak Al-Abdullah, Kuwait, 32093, Kuwait
- Adnan Kassar School of Business, Lebanese American University, Beirut, 03797751, Lebanon
| | - Chander Prakash
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, 140413, India
| | - Xiangdong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, 3000-548, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, Coimbra, 3000-548, Portugal
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
- University Centre for Research & Development, Chandigarh University, Mohali, Punjab, 140413, India
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, 600077, India
| | - Yi Xu
- Department of Science & Technology, Department of Urology, NanoBioMed Group, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| |
Collapse
|
7
|
Xin Z, Qu S, Qu Y, Xu Y, Liu R, Sun D, Dai Z. Emerging IL-12-based nanomedicine for cancer therapy. NANO TODAY 2024; 57:102331. [DOI: 10.1016/j.nantod.2024.102331] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
8
|
Leyden MC, Oviedo F, Saxena S, Kumar R, Le N, Reineke TM. Synergistic Polymer Blending Informs Efficient Terpolymer Design and Machine Learning Discerns Performance Trends for pDNA Delivery. Bioconjug Chem 2024; 35:897-911. [PMID: 38924453 DOI: 10.1021/acs.bioconjchem.4c00028] [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: 06/28/2024]
Abstract
Cationic polymers offer an alternative to viral vectors in nucleic acid delivery. However, the development of polymer vehicles capable of high transfection efficiency and minimal toxicity has remained elusive, and continued exploration of the vast design space is required. Traditional single polymer syntheses with large monomer bases are very time-intensive, limiting the speed at which new formulations are identified. In this work, we present an experimental method for the quick probing of the design space, utilizing a combinatorial set of 90 polymer blends, derived from 6 statistical copolymers, to deliver pDNA. This workflow facilitated rapid screening of polyplex compositions, successfully tailoring polyplex hydrophobicity, particle size, and payload binding affinity. This workflow identified blended polyplexes with high levels of transfection efficiency and cell viability relative to single copolymer controls and commercial JetPEI, indicating synergistic benefits from copolymer blending. Polyplex composition was coupled with biological outputs to guide the synthesis of single terpolymer vehicles, with high-performing polymers P10 and M20, providing superior transfection of HEK293T cells in serum-free and serum-containing media, respectively. Machine learning coupled with SHapley Additive exPlanations (SHAP) was used to identify polymer/polyplex attributes that most impact transfection efficiency, viability, and overall effective efficiency. Subsequent transfections on ARPE-19 and HDFn cells found that P10 and M20 were surpassed in performance by M10, contrasting with results in HEK293T cells. This cell type dependency reinforced the need to evaluate transfection conditions with multiple cell models to potentially identify moieties more beneficial to delivery in certain tissues. Overall, the workflow employed can be used to expedite the exploration of the polymer design space, bypassing extensive synthesis, and to develop improved polymer delivery vehicles more readily for nucleic acid therapies.
Collapse
Affiliation(s)
- Michael C Leyden
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Felipe Oviedo
- Nanite Inc., Boston, Massachusetts 02109, United States
| | - Sonashree Saxena
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ramya Kumar
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado 80401, United States
| | - Ngoc Le
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
9
|
Sadeghi M, Moslehi A, Kheiry H, Kiani FK, Zarei A, Khodakarami A, Karpisheh V, Masjedi A, Rahnama B, Hojjat-Farsangi M, Raeisi M, Yousefi M, Movasaghpour Akbari AA, Jadidi-Niaragh F. The sensitivity of acute myeloid leukemia cells to cytarabine is increased by suppressing the expression of Heme oxygenase-1 and hypoxia-inducible factor 1-alpha. Cancer Cell Int 2024; 24:217. [PMID: 38918761 PMCID: PMC11197338 DOI: 10.1186/s12935-024-03393-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 06/02/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML), a malignancy Often resistant to common chemotherapy regimens (Cytarabine (Ara-c) + Daunorubicin (DNR)), is accompanied by frequent relapses. Many factors are involved in causing chemoresistance. Heme Oxygenase-1 (HO-1) and Hypoxia-Inducible Factor 1-alpha (HIF-1α) are two of the most well-known genes, reported to be overexpressed in AML and promote resistance against chemotherapy according to several studies. The main chemotherapy agent used for AML treatment is Ara-c. We hypothesized that simultaneous targeting of HO-1 and HIF-1α could sensitize AML cells to Ara-c. METHOD In this study, we used our recently developed, Trans-Activator of Transcription (TAT) - Chitosan-Carboxymethyl Dextran (CCMD) - Poly Ethylene Glycol (PEG) - Nanoparticles (NPs), to deliver Ara-c along with siRNA molecules against the HO-1 and HIF-1α genes to AML primary cells (ex vivo) and cell lines including THP-1, KG-1, and HL-60 (in vitro). Subsequently, the effect of the single or combinational treatment on the growth, proliferation, apoptosis, and Reactive Oxygen Species (ROS) formation was evaluated. RESULTS The designed NPs had a high potential in transfecting cells with siRNAs and drug. The results demonstrated that treatment of cells with Ara-c elevated the generation of ROS in the cells while decreasing the proliferation potential. Following the silencing of HO-1, the rate of apoptosis and ROS generation in response to Ara-c increased significantly. While proliferation and growth inhibition were considerably evident in HIF-1α-siRNA-transfected-AML cells compared to cells treated with free Ara-c. We found that the co-inhibition of genes could further sensitize AML cells to Ara-c treatment. CONCLUSIONS As far as we are aware, this study is the first to simultaneously inhibit the HO-1 and HIF-1α genes in AML using NPs. It can be concluded that HO-1 causes chemoresistance by protecting cells from ROS damage. Whereas, HIF-1α mostly exerts prolific and direct anti-apoptotic effects. These findings imply that simultaneous inhibition of HO-1 and HIF-1α can overcome Ara-c resistance and help improve the prognosis of AML patients.
Collapse
Affiliation(s)
- Mohammad Sadeghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asma Moslehi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadiseh Kheiry
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Karoon Kiani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asieh Zarei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atefeh Khodakarami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Karpisheh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Masjedi
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Badrossadat Rahnama
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mortaza Raeisi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Tarvirdipour S, Skowicki M, Maffeis V, Abdollahi SN, Schoenenberger CA, Palivan CG. Peptide nanocarriers co-delivering an antisense oligonucleotide and photosensitizer elicit synergistic cytotoxicity. J Colloid Interface Sci 2024; 664:338-348. [PMID: 38479270 DOI: 10.1016/j.jcis.2024.03.021] [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: 12/18/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 04/07/2024]
Abstract
Combination therapies demand co-delivery platforms with efficient entrapment of distinct payloads and specific delivery to cells and possibly organelles. Herein, we introduce the combination of two therapeutic modalities, gene and photodynamic therapy, in a purely peptidic platform. The simultaneous formation and cargo loading of the multi-micellar platform is governed by self-assembly at the nanoscale. The multi-micellar architecture of the nanocarrier and the positive charge of its constituent micelles offer controlled dual loading capacity with distinct locations for a hydrophobic photosensitizer (PS) and negatively charged antisense oligonucleotides (ASOs). Moreover, the nuclear localization signal (NLS) sequence built-in the peptide targets PS + ASO-loaded nanocarriers to the nucleus. Breast cancer cells treated with nanocarriers demonstrated photo-triggered enhancement of radical oxygen species (ROS) associated with increased cell death. Besides, delivery of ASO payloads resulted in up to 90 % knockdown of Bcl-2, an inhibitor of apoptosis that is overexpressed in more than half of all human cancers. Simultaneous delivery of PS and ASO elicited synergistic apoptosis to an extent that could not be reached by singly loaded nanocarriers or the free form of the drugs. Both, the distinct location of loaded compounds that prevents them from interfering with each other, and the highly efficient cellular delivery support the great potential of this versatile peptide platform in combination therapy.
Collapse
Affiliation(s)
- Shabnam Tarvirdipour
- Department of Chemistry, University of Basel, Mattenstrasse 22, Basel 4058, Switzerland
| | - Michal Skowicki
- Department of Chemistry, University of Basel, Mattenstrasse 22, Basel 4058, Switzerland; NCCR-Molecular Systems Engineering, Mattenstrasse 24a, Basel 4058, Switzerland
| | - Viviana Maffeis
- Department of Chemistry, University of Basel, Mattenstrasse 22, Basel 4058, Switzerland; NCCR-Molecular Systems Engineering, Mattenstrasse 24a, Basel 4058, Switzerland
| | - S Narjes Abdollahi
- Department of Chemistry, University of Basel, Mattenstrasse 22, Basel 4058, Switzerland
| | | | - Cornelia G Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 22, Basel 4058, Switzerland; NCCR-Molecular Systems Engineering, Mattenstrasse 24a, Basel 4058, Switzerland.
| |
Collapse
|
11
|
Sandbhor P, Palkar P, Bhat S, John G, Goda JS. Nanomedicine as a multimodal therapeutic paradigm against cancer: on the way forward in advancing precision therapy. NANOSCALE 2024. [PMID: 38470224 DOI: 10.1039/d3nr06131k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Recent years have witnessed dramatic improvements in nanotechnology-based cancer therapeutics, and it continues to evolve from the use of conventional therapies (chemotherapy, surgery, and radiotherapy) to increasingly multi-complex approaches incorporating thermal energy-based tumor ablation (e.g. magnetic hyperthermia and photothermal therapy), dynamic therapy (e.g. photodynamic therapy), gene therapy, sonodynamic therapy (e.g. ultrasound), immunotherapy, and more recently real-time treatment efficacy monitoring (e.g. theranostic MRI-sensitive nanoparticles). Unlike monotherapy, these multimodal therapies (bimodal, i.e., a combination of two therapies, and trimodal, i.e., a combination of more than two therapies) incorporating nanoplatforms have tremendous potential to improve the tumor tissue penetration and retention of therapeutic agents through selective active/passive targeting effects. These combinatorial therapies can correspondingly alleviate drug response against hypoxic/acidic and immunosuppressive tumor microenvironments and promote/induce tumor cell death through various multi-mechanisms such as apoptosis, autophagy, and reactive oxygen-based cytotoxicity, e.g., ferroptosis, etc. These multi-faced approaches such as targeting the tumor vasculature, neoangiogenic vessels, drug-resistant cancer stem cells (CSCs), preventing intra/extravasation to reduce metastatic growth, and modulation of antitumor immune responses work complementary to each other, enhancing treatment efficacy. In this review, we discuss recent advances in different nanotechnology-mediated synergistic/additive combination therapies, emphasizing their underlying mechanisms for improving cancer prognosis and survival outcomes. Additionally, significant challenges such as CSCs, hypoxia, immunosuppression, and distant/local metastasis associated with therapy resistance and tumor recurrences are reviewed. Furthermore, to improve the clinical precision of these multimodal nanoplatforms in cancer treatment, their successful bench-to-clinic translation with controlled and localized drug-release kinetics, maximizing the therapeutic window while addressing safety and regulatory concerns are discussed. As we advance further, exploiting these strategies in clinically more relevant models such as patient-derived xenografts and 3D organoids will pave the way for the application of precision therapy.
Collapse
Affiliation(s)
- Puja Sandbhor
- Institute for NanoBioTechnology, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
| | - Pranoti Palkar
- Radiobiology, Department of Radiation Oncology & Homi Bhabha National Institute, Mumbai, 400012, India
| | - Sakshi Bhat
- Radiobiology, Department of Radiation Oncology & Homi Bhabha National Institute, Mumbai, 400012, India
| | - Geofrey John
- Radiobiology, Department of Radiation Oncology & Homi Bhabha National Institute, Mumbai, 400012, India
| | - Jayant S Goda
- Radiobiology, Department of Radiation Oncology & Homi Bhabha National Institute, Mumbai, 400012, India
| |
Collapse
|
12
|
Zhou LL, Guan Q, Dong YB. Covalent Organic Frameworks: Opportunities for Rational Materials Design in Cancer Therapy. Angew Chem Int Ed Engl 2024; 63:e202314763. [PMID: 37983842 DOI: 10.1002/anie.202314763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/18/2023] [Accepted: 11/20/2023] [Indexed: 11/22/2023]
Abstract
Nanomedicines are extensively used in cancer therapy. Covalent organic frameworks (COFs) are crystalline organic porous materials with several benefits for cancer therapy, including porosity, design flexibility, functionalizability, and biocompatibility. This review examines the use of COFs in cancer therapy from the perspective of reticular chemistry and function-oriented materials design. First, the modification sites and functionalization methods of COFs are discussed, followed by their potential as multifunctional nanoplatforms for tumor targeting, imaging, and therapy by integrating functional components. Finally, some challenges in the clinical translation of COFs are presented with the hope of promoting the development of COF-based anticancer nanomedicines and bringing COFs closer to clinical trials.
Collapse
Affiliation(s)
- Le-Le Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan, 250014, China
| | - Qun Guan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan, 250014, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau Taipa, Macau SAR, 999078, China
| | - Yu-Bin Dong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan, 250014, China
| |
Collapse
|
13
|
Brock SL, Badv M, Khademhosseini A, Weiss PS. Innovations in Bioengineering Virtual Special Issue. ACS MATERIALS AU 2023; 3:569-570. [PMID: 38089662 PMCID: PMC10636782 DOI: 10.1021/acsmaterialsau.3c00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Indexed: 10/13/2024]
|