1
|
Alsaidan OA. Recent advancements in aptamers as promising nanotool for therapeutic and diagnostic applications. Anal Biochem 2025; 702:115844. [PMID: 40090606 DOI: 10.1016/j.ab.2025.115844] [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: 12/17/2024] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/18/2025]
Abstract
Aptamers are single-strand oligonucleotide molecules having certain structural interactions which allow them to bind to specific targets. Modified nucleotides are added during or after a selection procedure like Systematic Evolution of Ligands by Exponential Enrichment i.e., SELEX to enhance the characteristics and functionality of aptamers. Aptamers are extensible molecular tools with several uses such as in drug administration, biosensing, bioimaging, drug therapies and diagnostics. The ability to detect is improved by using aptamer-based sensors in conjunction with biological molecules among other sensing techniques. Chemical modification, and strong resistance to denaturation, aptamers are appropriate biological recognizing agents for developing sensitive and repeatable aptasensors. This review discusses the most current developments in the aptamers, SELEX method, applications of aptamers as innovative diagnostic, therapeutic & theragnostic tool along with major limitations & prospective directions in the future.
Collapse
Affiliation(s)
- Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, P.O. Box 2014, Sakaka, 72341, Saudi Arabia.
| |
Collapse
|
2
|
Alavinejad M, Shirzad M, Javid-Naderi MJ, Rahdar A, Fathi-Karkan S, Pandey S. Smart nanomedicines powered by artificial intelligence: a breakthrough in lung cancer diagnosis and treatment. Med Oncol 2025; 42:134. [PMID: 40131617 DOI: 10.1007/s12032-025-02680-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
Lung cancer remains one of the leading causes of cancer-related mortality worldwide, primarily due to challenges in early detection, suboptimal therapeutic efficacy, and severe adverse effects associated with conventional treatments. The convergence of nanotechnology and artificial intelligence (AI) offers transformative potential in precision oncology, enabling innovative solutions for lung cancer diagnosis and therapy. Intelligent nanomedicines facilitate targeted drug delivery, enhanced imaging, and theranostic applications, while AI-driven models harness big biomedical data to optimize nanomedicine design, functionality, and clinical application. This review explores the synergistic integration of AI and nanotechnology in lung cancer care, highlighting recent advancements, key challenges, and future directions for clinical translation. Ethical considerations, including data standardization and privacy concerns, are also addressed, providing a comprehensive roadmap to overcome current barriers and advance the adoption of AI-driven intelligent nanomedicines in precision oncology. This synthesis underscores the critical role of emerging technologies in revolutionizing lung cancer management.
Collapse
Affiliation(s)
- Moloudosadat Alavinejad
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico, Universitat Politècnica de València, Universitat de València, Valencia, Spain
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València-Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Maryam Shirzad
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Javad Javid-Naderi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol, Iran.
| | - Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran.
- Department of Medical Nanotechnology, School of Medicine, North Khorasan University of Medical Science, Bojnurd, Iran.
| | - Sadanand Pandey
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India.
- Department of Chemistry, College of Natural Sciences, Yeungnam University, Gyeongsan, 38541, Gyeongbuk, Republic of Korea.
| |
Collapse
|
3
|
Helal MW, Faried MM, Salah SM, Ashraf M, Nasser N, Shawky Y, Hamdy S, Amir AE, Nabil W, El-Husseini DM. Comparative Analysis of Aptamer-Conjugated Chemical and Green Synthesized Gold Nanoparticles for Targeted Therapy in MCF-7 Cancer Cells. Appl Biochem Biotechnol 2025; 197:1678-1695. [PMID: 39601974 PMCID: PMC11953193 DOI: 10.1007/s12010-024-05091-2] [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] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
Breast cancer remains a challenging health issue, demanding innovative treatment approaches that maximize efficacy while minimizing damage to healthy cells. Targeted therapy offers a promising strategy tailored to the unique characteristics of breast cancer tumors. Gold nanoparticles have been studied in the context of their therapeutic potential towards cancer treatment showing great success. Recently, aptamers were also investigated for their targeting efficiency towards specific receptors allowing their use in targeting delivery systems. In this study, computational analysis was used to confirm the strong binding between AS1411 aptamer and the nucleolin receptor extensively present on the surface of breast cancer cells, highlighting the aptamer's potential for specific targeting. Furthermore, we investigated and compared the use of AS1411 aptamer-conjugated chemically synthesized (GNPs) and flaxseed-green-synthesized (Fs-GNPs) gold nanoparticles as targeting therapeutic systems for breast cancer cells. Our results showed successful conjugation of the AS1411 aptamer with both, the GNPs and Fs-GNPs. Characterization of the nanoparticles and their conjugates validates their size, charge, and morphology, affirming the success of the conjugation process. Cytotoxicity assessments using the MTT assay demonstrated the effectiveness of the conjugates against breast cancer cells, with the AS1411-Fs-GNPs conjugate exhibiting higher inhibitory efficacy, featuring an IC50 value of 11.13 µg/ml. In contrast, they showed minimal effect on normal cells, emphasizing the selectivity and potential safety of these therapies. To our knowledge, this is the first report of conjugating AS1411 aptamer to green-synthesized gold nanoparticles and its use as a targeting therapeutic system.
Collapse
Affiliation(s)
- Mariam W Helal
- Biotechnology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Mohanad M Faried
- Biotechnology Department, Faculty of Science, Cairo University, Giza, Egypt
| | | | - Mazen Ashraf
- Biotechnology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Nada Nasser
- Biotechnology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Yasser Shawky
- Biotechnology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Sara Hamdy
- Biotechnology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Azza El Amir
- Biotechnology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Wajeet Nabil
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Dalia M El-Husseini
- Nanomaterials Research and Synthesis Unit, Animal Health Research Institute (AHRI), Agricultural Research Center (ARC), Giza, Egypt.
| |
Collapse
|
4
|
Poudel K, Vithiananthan T, Kim JO, Tsao H. Recent progress in cancer vaccines and nanovaccines. Biomaterials 2025; 314:122856. [PMID: 39366184 DOI: 10.1016/j.biomaterials.2024.122856] [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: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
Vaccine science, nanotechnology, and immunotherapy are at the forefront of cancer treatment strategies, each offering significant potential for enhancing tumor-specific immunity and establishing long-lasting immune memory to prevent tumor recurrence. Despite the promise of these personalized and precision-based anti-cancer approaches, challenges such as immunosuppression, suboptimal immune activation, and T-cell exhaustion continue to hinder their effectiveness. The limited clinical success of cancer vaccines often stems from difficulties in identifying effective antigens, efficiently targeting immune cells, lymphoid organs, and the tumor microenvironment, overcoming immune evasion, enhancing immunogenicity, and avoiding lysosomal degradation. However, numerous studies have demonstrated that integrating nanotechnology with immunotherapeutic strategies in vaccine development can overcome these challenges, leading to potent antitumor immune responses and significant progress in the field. This review highlights the critical components of cancer vaccine and nanovaccine strategies for immunomodulatory antitumor therapy. It covers general vaccine strategies, types of vaccines, antigen forms, nanovaccine platforms, challenges faced, potential solutions, and key findings from preclinical and clinical studies, along with future perspectives. To fully unlock the potential of cancer vaccines and nanovaccines, precise immunological monitoring during early-phase trials is essential. This approach will help identify and address obstacles, ultimately expanding the available options for patients who are resistant to conventional cancer immunotherapies.
Collapse
Affiliation(s)
- Kishwor Poudel
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tulasi Vithiananthan
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Ebrahimi F, Kumari A, Al Abdullah S, Vivero-Escoto JL, Dellinger K. Surface Functionalization of Citrate-Stabilized Gold Nanoparticles with Various Disease-Specific Nonthiolated Aptamers: RSM-Based Optimization for Multifactorial Disease Biomarker Detection. ACS Sens 2025; 10:944-953. [PMID: 39960422 DOI: 10.1021/acssensors.4c02722] [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: 03/01/2025]
Abstract
This study focuses on the surface functionalization of citrate-stabilized gold nanoparticles (AuNPs) with disease-specific aptamers to enhance the detection of multifactorial disease (MD) biomarkers. MDs, characterized by complex pathophysiology involving multiple genetic and environmental factors, present significant diagnostic challenges. Aptamers, which are short, single-stranded oligonucleotides with high specificity and affinity for target molecules, have emerged as promising tools for biomarker detection. By utilizing response surface methodology (RSM) and face-centered central composite design (FCCCD), this research systematically optimized the bioconjugation process of AuNPs with different aptamer sequences, focusing on parameters such as AuNP size and aptamer concentration. The developed protocol in this study demonstrated that aptamer-functionalized AuNPs can be optimized for high yield, bioconjugation efficiency, stability, and surface coverage, making them suitable for diagnostic applications, particularly in surface-enhanced Raman spectroscopy (SERS). The findings provide a foundation for the development of customizable nanoprobes that can be adapted for the detection of various biomarkers associated with MDs, potentially improving early diagnosis and therapeutic outcomes.
Collapse
Affiliation(s)
- Farbod Ebrahimi
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 East Gate City Boulevard, Greensboro, North Carolina 27401, United States
| | - Anjali Kumari
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 East Gate City Boulevard, Greensboro, North Carolina 27401, United States
| | - Saqer Al Abdullah
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 East Gate City Boulevard, Greensboro, North Carolina 27401, United States
| | - Juan L Vivero-Escoto
- Department of Chemistry, University of North Carolina at Charlotte, 9201 University City Boulevard, Charlotte, North Carolina 28223, United States
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 East Gate City Boulevard, Greensboro, North Carolina 27401, United States
| |
Collapse
|
6
|
Zhao R, Lan D, Xia B, Dong M, Mu J, Zhao Y. PET-Based Dual-Modal Probes for In Vivo Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409713. [PMID: 39873346 DOI: 10.1002/smll.202409713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/07/2024] [Indexed: 01/30/2025]
Abstract
Molecular imaging has significantly advanced the detection and analysis of in vivo metabolic processes, while single-modal techniques remain limited. Dual-modal imaging, particularly positron emission tomography (PET)-based combinations has emerged as a powerful solution, offering enhanced capabilities through integration with magnetic resonance imaging (MRI) or near-infrared fluorescence (NIRF) imaging. This review highlights recent progress in PET-based dual-modal imaging, focusing on the development of various bimodal probes derived from antibodies, nanoparticles, and peptides, and key applications including image-guided surgery and disease assessment. PET-based dual-modal imaging holds substantial potential for advancing research and diagnostics by improving resolution and providing functional insights. By combining complementary modalities, these systems deliver a more comprehensive view of disease processes, leading to more accurate diagnoses and targeted treatments. Future research prioritizes optimizing probe design for enhanced biocompatibility and safety, facilitating clinical translation, and broadens applications beyond cancer. Through interdisciplinary collaboration, PET-based dual-modal probes are poised to play a pivotal role in improving patient outcomes, particularly in diagnosing and managing complex diseases.
Collapse
Affiliation(s)
- Runge Zhao
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Deren Lan
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Beibei Xia
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - MengJie Dong
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Jing Mu
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Yongsheng Zhao
- Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| |
Collapse
|
7
|
Sandeep, Subba R, Mondal AC. Does COVID-19 Trigger the Risk for the Development of Parkinson's Disease? Therapeutic Potential of Vitamin C. Mol Neurobiol 2024; 61:9945-9960. [PMID: 37957424 DOI: 10.1007/s12035-023-03756-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), which was proclaimed a pandemic by the World Health Organization (WHO) in March 2020. There is mounting evidence that older patients with multimorbidity are more susceptible to COVID-19 complications than are younger, healthy people. Having neuroinvasive potential, SARS-CoV-2 infection may increase susceptibility toward the development of Parkinson's disease (PD), a progressive neurodegenerative disorder with extensive motor deficits. PD is characterized by the aggregation of α-synuclein in the form of Lewy bodies and the loss of dopaminergic neurons in the dorsal striatum and substantia nigra pars compacta (SNpc) of the nigrostriatal pathway in the brain. Increasing reports suggest that SARS-CoV-2 infection is linked with the worsening of motor and non-motor symptoms with high rates of hospitalization and mortality in PD patients. Common pathological changes in both diseases involve oxidative stress, mitochondrial dysfunction, neuroinflammation, and neurodegeneration. COVID-19 exacerbates the damage ensuing from the dysregulation of those processes, furthering neurological complications, and increasing the severity of PD symptomatology. Phytochemicals have antioxidant, anti-inflammatory, and anti-apoptotic properties. Vitamin C supplementation is found to ameliorate the common pathological changes in both diseases to some extent. This review aims to present the available evidence on the association between COVID-19 and PD, and discusses the therapeutic potential of vitamin C for its better management.
Collapse
Affiliation(s)
- Sandeep
- Laboratory of Cellular & Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Rhea Subba
- Laboratory of Cellular & Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Amal Chandra Mondal
- Laboratory of Cellular & Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
8
|
Handali S, Rezaei M. Aptamer-decorated nanocarriers for viral adsorption: A special look at COVID-19. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102310. [PMID: 39281706 PMCID: PMC11401170 DOI: 10.1016/j.omtn.2024.102310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Viral infections are one of the leading causes of death in the world. One main challenge in fighting against these diseases is the unavailability of effective eradicating drugs and specific treatments. Nanocarriers and aptamer-decorated nanocarriers are designed to attach to many targets, including viral particles. By lowering the viral infectivity and attachment capability, they add therapeutic values even without containing antiviral drugs. Nevertheless, the nanoparticles (NPs) with encapsulated antiviral drugs can display extra therapeutic effects. Furthermore, it has been shown that aptamers can bind to viral particles and nanocarriers, presenting promising approaches for the identification of viruses and treatment of viral infections. Although there is no satisfying literature revealing the strong therapeutic potential of nanotechnology against COVID-19, the following information can provide new perspectives for upcoming investigations pertaining to developing effective aptamer-nanocarrier agents against COVID-19.
Collapse
Affiliation(s)
- Somayeh Handali
- Medical Biomaterials Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Rezaei
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Institute for Natural Products and Medicinal Plants (INPMP), Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
9
|
Yu H, Liu S, Yuan Z, Huang H, Yan P, Zhu W. Targeted co-delivery of rapamycin and oxaliplatin by liposomes suppresses tumor growth and metastasis of colorectal cancer. Biomed Pharmacother 2024; 178:117192. [PMID: 39098178 DOI: 10.1016/j.biopha.2024.117192] [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: 04/30/2024] [Revised: 07/08/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024] Open
Abstract
The activation of tumor cell immunogenicity through oxaliplatin (OXP)-induced immunogenic cell death (ICD) has significant implications in cancer treatment. However, the anti-tumor effect of OXP monotherapy still has many shortcomings, and the systemic administration of OXP leads to low drug concentration at the tumor site, which is susceptible to systemic toxic side effects. In this study, a combined therapeutic strategy using folate-modified nanoliposomes co-delivered with rapamycin (Rapa) and OXP (abbreviated as FA@R/O Lps) is proposed for the treatment of colorectal cancer (CRC). Rapa and OXP can directly inhibit tumor cell proliferation and induce apoptosis. OXP induces ICD by triggering the release of danger signals, such as HMGB1, ATP, and calreticulin. FA@R/O Lps with a particle size of about 134.1±1.8 nm and a small dispersion were successfully prepared. This novel liposomal system can be used to target and increase drug accumulation in tumors. In-vivo experiments showed that FA@R/O Lps successfully inhibit CRC growth and liver metastasis, and simultaneously reduce off-target toxicity. In particular, FA@R/O Lps showed greater therapeutic effects than free Rapa/OXP and R/O Lps. Taken together, this study provides a novel combination of Rapa and OXP, and a nano-delivery system for enhanced anti-CRC efficacy. The results suggest that FA@R/O Lps could be a promising strategy for the treatment of CRC.
Collapse
Affiliation(s)
- Hang Yu
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Shengyao Liu
- Department of Spinal Surgery, The Second Affiliated Hospital of Guangzhou Medical University, No. 250, Changgangdong Road, Guangzhou 510260, China
| | - Zhongwen Yuan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Hanhui Huang
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Pengke Yan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| | - Wenting Zhu
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| |
Collapse
|
10
|
Park SG, Lee HJ, Ji T, Kim K, Ohk SH. Aptamer Based SPREETA Sensor for the Detection of Porphyromonas gingivalis G-Protein. J Microbiol Biotechnol 2024; 34:289-295. [PMID: 38111313 PMCID: PMC10940744 DOI: 10.4014/jmb.2310.10042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/20/2023]
Abstract
We have developed an aptamer that specifically binds to Porphyromonas gingivalis to reduce the cellular damage caused by P. gingivalis infection and applied it as a biosensor. P. gingivalis is one of the major pathogens causing destructive periodontal disease among the periodontal microorganisms constituting complex biofilms. Porphyromonas gingivalis G-protein (PGP) known to play an important role in the transmission of germs was used as a target protein for the screening of aptamer. The aptamer that has binds to the G-protein of P. gingivalis, was screened and developed through the Systemic Evolution of Ligands by Exponential Energy (SELEX) method. Modified-Western blot analysis was performed with the aptamer which consisted of 38 single-stranded DNA to confirm the selectivity. ELONA (enzyme linked oligonucleotide assay) used to confirm that the aptamer was sensitive to PGP even at low concentration of 1 μg/ml. For the rapid detection of P. gingivalis, we constructed a surface plasmon resonance biosensor with SPREETA using the PGP aptamer. It was confirmed that PGP could be detected as low concentration as at 0.1 pM, which is the minimum concentration of aptamer sensor within 5 min. Based on these results, we have constructed a SPREETA biosensor based on aptamer that can bind to P. gingivalis G-protein. It can be used as an infection diagnosis system to rapidly diagnose and analyze oral diseases caused by P. gingivalis.
Collapse
Affiliation(s)
- Suk-Gyun Park
- Department of Oral Microbiology, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hyun Ju Lee
- Department of Cosmetic Science, Kwangju Women’s University, Gwangju 62396, Republic of Korea
| | - Taeksoo Ji
- School of Electronics and Computer Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kyungbaek Kim
- School of Electronics and Computer Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seung-Ho Ohk
- Department of Oral Microbiology, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
11
|
Ghadin N, Yusof NAM, Syarul Nataqain B, Raston NHA, Low CF. Selection and characterization of ssDNA aptamer targeting Macrobrachium rosenbergii nodavirus capsid protein: A potential capture agent in gold-nanoparticle-based aptasensor for viral protein detection. JOURNAL OF FISH DISEASES 2024; 47:e13892. [PMID: 38014615 DOI: 10.1111/jfd.13892] [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: 08/23/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
The giant freshwater prawn holds a significant position as a valuable crustacean species cultivated in the aquaculture industry, particularly well-known and demanded among the Southeast Asian countries. Aquaculture production of this species has been impacted by Macrobrachium rosenbergii nodavirus (MrNV) infection, which particularly affects the larvae and post-larvae stages of the prawn. The infection has been recorded to cause mortality rates of up to 100% among the affected prawns. A simple, fast, and easy to deploy on-site detection or diagnostic method is crucial for early detection of MrNV to control the disease outbreak. In the present study, novel single-stranded DNA aptamers targeting the MrNV capsid protein were identified using the systematic evolution of ligands by exponential enrichment (SELEX) approach. The aptamer was then conjugated with the citrate-capped gold nanoparticles (AuNPs), and the sensitivity of this AuNP-based aptasensor for the detection of MrNV capsid protein was evaluated. Findings revealed that the aptamer candidate, APT-MrNV-CP-1 was enriched throughout the SELEX cycle 4, 9, and 12 with the sequence percentage of 1.76%, 9.09%, and 12.42%, respectively. The conjugation of APT-MrNV-CP-1 with citrate-capped AuNPs exhibited the highest sensitivity in detecting the MrNV capsid protein, where the presence of 62.5 nM of the viral capsid protein led to a significant agglomeration of the AuNPs. This study demonstrated the practicality of an AuNP-based aptasensor for disease diagnosis, particularly for detecting MrNV infection in giant freshwater prawns.
Collapse
Affiliation(s)
- Norazli Ghadin
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Nur Afiqah Md Yusof
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | | | - Nurul Hanun Ahmad Raston
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Chen Fei Low
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| |
Collapse
|
12
|
Gurunathan S, Thangaraj P, Wang L, Cao Q, Kim JH. Nanovaccines: An effective therapeutic approach for cancer therapy. Biomed Pharmacother 2024; 170:115992. [PMID: 38070247 DOI: 10.1016/j.biopha.2023.115992] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Cancer vaccines hold considerable promise for the immunotherapy of solid tumors. Nanomedicine offers several strategies for enhancing vaccine effectiveness. In particular, molecular or (sub) cellular vaccines can be delivered to the target lymphoid tissues and cells by nanocarriers and nanoplatforms to increase the potency and durability of antitumor immunity and minimize negative side effects. Nanovaccines use nanoparticles (NPs) as carriers and/or adjuvants, offering the advantages of optimal nanoscale size, high stability, ample antigen loading, high immunogenicity, tunable antigen presentation, increased retention in lymph nodes, and immunity promotion. To induce antitumor immunity, cancer vaccines rely on tumor antigens, which are administered in the form of entire cells, peptides, nucleic acids, extracellular vesicles (EVs), or cell membrane-encapsulated NPs. Ideal cancer vaccines stimulate both humoral and cellular immunity while overcoming tumor-induced immune suppression. Herein, we review the key properties of nanovaccines for cancer immunotherapy and highlight the recent advances in their development based on the structure and composition of various (including synthetic and semi (biogenic) nanocarriers. Moreover, we discuss tumor cell-derived vaccines (including those based on whole-tumor-cell components, EVs, cell membrane-encapsulated NPs, and hybrid membrane-coated NPs), nanovaccine action mechanisms, and the challenges of immunocancer therapy and their translation to clinical applications.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641 021, Tamil Nadu, India.
| | - Pratheep Thangaraj
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641 021, Tamil Nadu, India
| | - Lin Wang
- Research and Development Department, Qingdao Haier Biotech Co., Ltd., Qingdao, China
| | - Qilong Cao
- Research and Development Department, Qingdao Haier Biotech Co., Ltd., Qingdao, China
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
13
|
Liu MS, Zhong SS, Jiang S, Wang T, Zhang KH. Bibliometric analysis of aptamer-conjugated nanoparticles for diagnosis in the last two decades. NANOTECHNOLOGY 2023; 35:055102. [PMID: 37879319 DOI: 10.1088/1361-6528/ad06d5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/25/2023] [Indexed: 10/27/2023]
Abstract
Objective.Aptamer-conjugated nanoparticles for diagnosis have recently gained increasing attention. Here, we performed a bibliometric analysis to provide an overview of this field over the past two decades.Methods. The terms 'aptamer, nanoparticles and diagnosis' were used to search for relevant original articles published in English from 2003 to 2022 in the Web of Science database. VOSviewer and CiteSpace software were employed to analyze the development process, knowledge structure, research hotspots, and potential trends in the field of aptamer-conjugated nanoparticles for diagnosis.Results. A total of 1076 original articles were retrieved, with a rapid increase in the annual output and citation. The journal 'Biosensors and Bioelectronics' has contributed the most in this field, and the most influential researcher, institution and country were Weihong Tan, the Chinese Academy of Sciences, China, respectively. Gold nanoparticles and quantum dots were the most used, but in the past three years, research hotspots focused on carbon dots and graphene quantum dots. Diagnostic directions primarily focused on cancer. The most used strategy was label-free electrochemical detection, but in the past two years, colorimetric analysis and fluorescence imaging emerged as hot topics.Conclusion.The bibliometric analysis reveals a rapid increase in the research on aptamer-conjugated nanoparticles for diagnosis, major contributors at the levels of journals, authors, institutions, and countries, and research preferences in diagnostic objects, nanoparticle types, and detection methods, as well as the evolution of research hotspots and future trends.
Collapse
Affiliation(s)
- Mao-Sheng Liu
- Department of Gastroenterology, Jiangxi Institute of Gastroenterology & Hepatology, the First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Si-Si Zhong
- Department of Quality and Safety Management, the First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| | - Song Jiang
- Department of Gastroenterology, Jiangxi Institute of Gastroenterology & Hepatology, the First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Ting Wang
- Department of Gastroenterology, Jiangxi Institute of Gastroenterology & Hepatology, the First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Kun-He Zhang
- Department of Gastroenterology, Jiangxi Institute of Gastroenterology & Hepatology, the First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
14
|
Szymanowski W, Szymanowska A, Bielawska A, Lopez-Berestein G, Rodriguez-Aguayo C, Amero P. Aptamers as Potential Therapeutic Tools for Ovarian Cancer: Advancements and Challenges. Cancers (Basel) 2023; 15:5300. [PMID: 37958473 PMCID: PMC10647731 DOI: 10.3390/cancers15215300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Ovarian cancer (OC) is the most common lethal gynecologic cause of death in women worldwide, with a high mortality rate and increasing incidence. Despite advancements in the treatment, most OC patients still die from their disease due to late-stage diagnosis, the lack of effective diagnostic methods, and relapses. Aptamers, synthetic, short single-stranded oligonucleotides, have emerged as promising anticancer therapeutics. Their ability to selectively bind to target molecules, including cancer-related proteins and receptors, has revolutionized drug discovery and biomarker identification. Aptamers offer unique insights into the molecular pathways involved in cancer development and progression. Moreover, they show immense potential as drug delivery systems, enabling targeted delivery of therapeutic agents to cancer cells while minimizing off-target effects and reducing systemic toxicity. In the context of OC, the integration of aptamers with non-coding RNAs (ncRNAs) presents an opportunity for precise and efficient gene targeting. Additionally, the conjugation of aptamers with nanoparticles allows for accurate and targeted delivery of ncRNAs to specific cells, tissues, or organs. In this review, we will summarize the potential use and challenges associated with the use of aptamers alone or aptamer-ncRNA conjugates, nanoparticles, and multivalent aptamer-based therapeutics for the treatment of OC.
Collapse
Affiliation(s)
- Wojciech Szymanowski
- Department of Biotechnology, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.S.); (A.B.)
| | - Anna Szymanowska
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (G.L.-B.); (C.R.-A.)
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.S.); (A.B.)
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (G.L.-B.); (C.R.-A.)
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (G.L.-B.); (C.R.-A.)
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (G.L.-B.); (C.R.-A.)
| |
Collapse
|
15
|
Phalake SS, Somvanshi SB, Tofail SAM, Thorat ND, Khot VM. Functionalized manganese iron oxide nanoparticles: a dual potential magneto-chemotherapeutic cargo in a 3D breast cancer model. NANOSCALE 2023; 15:15686-15699. [PMID: 37724853 DOI: 10.1039/d3nr02816j] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
Localized heat generation from manganese iron oxide nanoparticles (MIONPs) conjugated with chemotherapeutics under the exposure of an alternating magnetic field (magneto-chemotherapy) can revolutionize targeted breast cancer therapy. On the other hand, the lack of precise control of local temperature and adequate MIONP distribution in laboratory settings using the conventional two-dimensional (2D) cellular models has limited its further translation in tumor sites. Our current study explored advanced 3D in vitro tumor models as a promising alternative to replicate the complete range of tumor characteristics. Specifically, we have focused on investigating the effectiveness of MIONP-based magneto-chemotherapy (MCT) as an anticancer treatment in a 3D breast cancer model. To achieve this, chitosan-coated MIONPs (CS-MIONPs) are synthesized and functionalized with an anticancer drug (doxorubicin) and a tumor-targeting aptamer (AS1411). CS-MIONPs with a crystallite size of 16.88 nm and a specific absorption rate (SAR) of 181.48 W g-1 are reported. In vitro assessment of MCF-7 breast cancer cell lines in 2D and 3D cell cultures demonstrated anticancer activity. In the 2D and 3D cancer models, the MIONP-mediated MCT reduced cancer cell viability to about 71.48% and 92.2%, respectively. On the other hand, MIONP-mediated MCT under an AC magnetic field diminished spheroids' viability to 83.76 ± 2%, being the most promising therapeutic modality against breast cancer.
Collapse
Affiliation(s)
- Satish S Phalake
- Department of Medical Physics, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416 006, Maharashtra, India.
| | - Sandeep B Somvanshi
- School of Materials Engineering, Purdue University, West Lafayette, USA
- Department of Physics, Dr. B. A. M. University, Aurangabad-431004, Maharashtra, India
| | - Syed A M Tofail
- Department of Physics and Bernal Institute, Limerick Digital Cancer Research Centre (LDCRC), University of Limerick, Castletroy, Co. Limerick, Limerick, V94 T9PX, Ireland.
| | - Nanasaheb D Thorat
- Department of Physics and Bernal Institute, Limerick Digital Cancer Research Centre (LDCRC), University of Limerick, Castletroy, Co. Limerick, Limerick, V94 T9PX, Ireland.
- Nuffield Department of Women's and Reproductive Health, John Radcliffe Hospital, Medical Sciences Division, University of Oxford, Oxford OX3 9DU, UK.
| | - Vishwajeet M Khot
- Department of Medical Physics, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Deemed to be University), Kolhapur, 416 006, Maharashtra, India.
| |
Collapse
|
16
|
Gupta J, Ahmed AT, Tayyib NA, Zabibah RS, Shomurodov Q, Kadheim MN, Alsaikhan F, Ramaiah P, Chinnasamy L, Samarghandian S. A state-of-art of underlying molecular mechanisms and pharmacological interventions/nanotherapeutics for cisplatin resistance in gastric cancer. Biomed Pharmacother 2023; 166:115337. [PMID: 37659203 DOI: 10.1016/j.biopha.2023.115337] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 09/04/2023] Open
Abstract
The fourth common reason of death among patients is gastric cancer (GC) and it is a dominant tumor type in Ease Asia. One of the problems in GC therapy is chemoresistance. Cisplatin (CP) is a platinum compound that causes DNA damage in reducing tumor progression and viability of cancer cells. However, due to hyperactivation of drug efflux pumps, dysregulation of genes and interactions in tumor microenvironment, tumor cells can develop resistance to CP chemotherapy. The current review focuses on the CP resistance emergence in GC cells with emphasizing on molecular pathways, pharmacological compounds for reversing chemoresistance and the role of nanostructures. Changes in cell death mechanisms such as upregulation of pro-survival autophagy can prevent CP-mediated apoptosis that results in drug resistance. Moreover, increase in metastasis via EMT induction induces CP resistance. Dysregulation of molecular pathways such as PTEN, PI3K/Akt, Nrf2 and others result in changes in CP response of GC cells. Non-coding RNAs determine CP response of GC cells and application of pharmacological compounds with activity distinct of CP can result in sensitivity in tumor cells. Due to efficacy of exosomes in transferring bioactive molecules such as RNA and DNA molecules among GC cells, exosomes can also result in CP resistance. One of the newest progresses in overcoming CP resistance in GC is application of nanoplatforms for delivery of CP in GC therapy that they can increase accumulation of CP at tumor site and by suppressing carcinogenic factors and overcoming biological barriers, they increase CP toxicity on cancer cells.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, U.P., India
| | | | - Nahla A Tayyib
- Faculty of Nursing, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Qakhramon Shomurodov
- Department of Maxillofacial Surgery, Tashkent State Dental Institute, Tashkent, Uzbekistan; Department of Scientific Affairs, Samarkand State Medical University, Samarkand, Uzbekistan
| | - Mostafai N Kadheim
- Department of Dentistry, Kut University College, Kut, Wasit 52001, Iraq; Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad 10022 Iraq
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| | | | | | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, the Islamic Republic of Iran.
| |
Collapse
|
17
|
Maki MAA, Teng MS, Tan KF, Kumar PV. Polyamidoamine-stabilized and hyaluronic acid-functionalized gold nanoparticles for cancer therapy. OPENNANO 2023; 13:100182. [DOI: 10.1016/j.onano.2023.100182] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
18
|
Jelen Ž, Kovač J, Rudolf R. Study of Gold Nanoparticles Conjugated with SARS-CoV-2 S1 Spike Protein Fragments. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2160. [PMID: 37570478 PMCID: PMC10421057 DOI: 10.3390/nano13152160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023]
Abstract
This study reports on the successful conjugation of SARS-CoV-2 S1 spike protein fragments with gold nanoparticles (AuNPs) that were synthesised with Ultrasonic Spray Pyrolysis (USP). This method enables the continuous synthesis of AuNPs with a high degree of purity, round shapes, and the formation of a surface that allows various modifications. The conjugation mechanism of USP synthesized AuNPs with SARS-CoV-2 S1 spike protein fragments was investigated. A gel electrophoresis experiment confirmed the successful conjugation of AuNPs with SARS-CoV-2 S1 fragments indirectly. X-ray Photoelectron Spectroscopy (XPS) analysis confirmed the presence of characteristic O1s and N1s peaks, which indicated that specific binding between AuNPs and SARS-CoV-2 S1 spike protein fragments takes place via a peptide bond formed with the citrate stabiliser. This bond is coordinated to the AuNP's surface and the N-terminals of the protein, with the conjugate displaying the expected response within a prototype LFIA test. This study will help in better understanding the behaviour of AuNPs synthesised with USP and their potential use as sensors in colorimetric or electrochemical sensors and LFIA tests.
Collapse
Affiliation(s)
- Žiga Jelen
- Faculty of Mechanical Engineering, University of Maribor, Smetanova ulica 17, SI-2000 Maribor, Slovenia;
| | - Janez Kovač
- Department of Surface Engineering, Institut Jožef Stefan, Jamova 39, SI-1000 Ljubljana, Slovenia;
| | - Rebeka Rudolf
- Faculty of Mechanical Engineering, University of Maribor, Smetanova ulica 17, SI-2000 Maribor, Slovenia;
| |
Collapse
|
19
|
Sultana R, Yadav D, Puranik N, Chavda V, Kim J, Song M. A Review on the Use of Gold Nanoparticles in Cancer Treatment. Anticancer Agents Med Chem 2023; 23:2171-2182. [PMID: 37842886 DOI: 10.2174/0118715206268664231004040210] [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: 07/17/2023] [Revised: 08/23/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023]
Abstract
According to a 2020 WHO study, cancer is responsible for one in every six fatalities. One in four patients die due to side effects and intolerance to chemotherapy, making it a leading cause of patient death. Compared to traditional tumor therapy, emerging treatment methods, including immunotherapy, gene therapy, photothermal therapy, and photodynamic therapy, have proven to be more effective. The aim of this review is to highlight the role of gold nanoparticles in advanced cancer treatment. A systematic and extensive literature review was conducted using the Web of Science, PubMed, EMBASE, Google Scholar, NCBI, and various websites. Highly relevant literature from 141 references was chosen for inclusion in this review. Recently, the synergistic benefits of nano therapy and cancer immunotherapy have been shown, which could allow earlier diagnosis, more focused cancer treatment, and improved disease control. Compared to other nanoparticles, the physical and optical characteristics of gold nanoparticles appear to have significantly greater effects on the target. It has a crucial role in acting as a drug carrier, biomarker, anti-angiogenesis agent, diagnostic agent, radiosensitizer, cancer immunotherapy, photodynamic therapy, and photothermal therapy. Gold nanoparticle-based cancer treatments can greatly reduce current drug and chemotherapy dosages.
Collapse
Affiliation(s)
- Razia Sultana
- Department of Zoology, SKM Govt College, Nawapara, Raipur, 493881, India
| | - Dhananjay Yadav
- Department of Life Sciences, Yeungnam University, 38541, Gyeongsan, Republic of Korea
| | - Nidhi Puranik
- Department of Biochemistry & Genetics, Barkatullah University, Bhopal, 462026, India
| | - Vishal Chavda
- Department of Pathology, Stanford School of Medicine, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - Jeongyeon Kim
- Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, 38541, Gyeongsan, Republic of Korea
| |
Collapse
|
20
|
Zhu Y, Yang D, Guo T, Lin M. Use of S2.2/DOX Magnetic Nanoliposomes in MR Molecule Imaging and Targeted Thermochemotherapy for Breast Cancer In Vitro. Technol Cancer Res Treat 2023; 22:15330338231194498. [PMID: 37563954 PMCID: PMC10422896 DOI: 10.1177/15330338231194498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE To prepare S2.2/DOX magnetic nanoliposomes by combining the potential benefits of MNPs in MRI and the targeted performance of nano-drugs as an innovative method for integrated diagnosis and treatment of breast cancer (BC). METHODS We created a S2.2-PEG-MZF/DOX molecular probe by using a lipid material to encapsulate PEG-MZF-NPs and doxorubicin (DOX), and a S2.2 aptamer to target MUC1 to conjugate with PEG-MZF/DOX nanoliposomes. The potential of probe for cell-specific targeting and magnetic resonance (MR) molecular imaging was evaluated by MR scanner and Prussian blue staining. Additionally, we explored the feasibility by using nanoliposome magnetic induction heating to interfere with MCF-7 (MUC1+) BC cells under the influence of an alternating magnetic field (AMF). RESULTS PEG-MZF-NPs were biologically safe. The T2 relaxation rate of PEG-MZF-NPs was found to inhibit T2 signal in a concentration-dependent manner, and the T2 signal of the S2.2-PEG-MZF molecular probe in MCF-7 cells was significantly lower than that in PEG-MZF-NPs group. Moreover, the T2 signal reduction was more pronounced in MCF-7 cells than in the hepatoma cell line HepG2 (MUC1-), suggesting a strong MRI potential of the S2.2-PEG-MZF molecular probe. The S2.2-PEG-MZF/DOX nanoliposome was able to achieve the desired temperature range for tumor hyperthermia (42-44 °C) in vitro. The S2.2-PEG-MZF/DOX nanoliposome accompanied by magnetic fluid hyperthermia (MFH) could inhibit proliferation and invasion and induce apoptosis of MCF-7 cells. The effects of this approach were significantly higher than those observed in the other groups. CONCLUSION We successfully developed a novel technique for BC diagnosis and treatment using thermochemotherapy under the guidance of MR molecular imaging. This approach holds great potential for improving the management of this devastating disease in the future.
Collapse
Affiliation(s)
- Yinxing Zhu
- Taizhou School of Clinical Medicine, Nanjing Medical University, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Dazhuang Yang
- Imaging Department, General Hospital of Xuzhou Mining Group, Xuzhou, Jiangsu, China
| | - Ting Guo
- Institute of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Mei Lin
- Clinical Laboratory, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| |
Collapse
|