1
|
Kumar S, Mohan A, Sharma NR, Kumar A, Girdhar M, Malik T, Verma AK. Computational Frontiers in Aptamer-Based Nanomedicine for Precision Therapeutics: A Comprehensive Review. ACS OMEGA 2024; 9:26838-26862. [PMID: 38947800 PMCID: PMC11209897 DOI: 10.1021/acsomega.4c02466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/09/2024] [Accepted: 05/28/2024] [Indexed: 07/02/2024]
Abstract
In the rapidly evolving landscape of nanomedicine, aptamers have emerged as powerful molecular tools, demonstrating immense potential in targeted therapeutics, diagnostics, and drug delivery systems. This paper explores the computational features of aptamers in nanomedicine, highlighting their advantages over antibodies, including selectivity, low immunogenicity, and a simple production process. A comprehensive overview of the aptamer development process, specifically the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) process, sheds light on the intricate methodologies behind aptamer selection. The historical evolution of aptamers and their diverse applications in nanomedicine are discussed, emphasizing their pivotal role in targeted drug delivery, precision medicine and therapeutics. Furthermore, we explore the integration of artificial intelligence (AI), machine learning (ML), Internet of Things (IoT), Internet of Medical Things (IoMT), and nanotechnology in aptameric development, illustrating how these cutting-edge technologies are revolutionizing the selection and optimization of aptamers for tailored biomedical applications. This paper also discusses challenges in computational methods for advancing aptamers, including reliable prediction models, extensive data analysis, and multiomics data incorporation. It also addresses ethical concerns and restrictions related to AI and IoT use in aptamer research. The paper examines progress in computer simulations for nanomedicine. By elucidating the importance of aptamers, understanding their superiority over antibodies, and exploring the historical context and challenges, this review serves as a valuable resource for researchers and practitioners aiming to harness the full potential of aptamers in the rapidly evolving field of nanomedicine.
Collapse
Affiliation(s)
- Shubham Kumar
- School
of Bioengineering and Biosciences, Lovely
Professional University, Phagwara, Punjab 144001, India
| | - Anand Mohan
- School
of Bioengineering and Biosciences, Lovely
Professional University, Phagwara, Punjab 144001, India
| | - Neeta Raj Sharma
- School
of Bioengineering and Biosciences, Lovely
Professional University, Phagwara, Punjab 144001, India
| | - Anil Kumar
- Gene
Regulation Laboratory, National Institute
of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Madhuri Girdhar
- Division
of Research and Development, Lovely Professional
University, Phagwara 144401, Punjab, India
| | - Tabarak Malik
- Department
of Biomedical Sciences, Institute of Health, Jimma University, MVJ4+R95 Jimma, Ethiopia
| | - Awadhesh Kumar Verma
- School
of Bioengineering and Biosciences, Lovely
Professional University, Phagwara, Punjab 144001, India
| |
Collapse
|
2
|
Ouyang Y, O'Hagan MP, Willner B, Willner I. Aptamer-Modified Homogeneous Catalysts, Heterogenous Nanoparticle Catalysts, and Photocatalysts: Functional "Nucleoapzymes", "Aptananozymes", and "Photoaptazymes". ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2210885. [PMID: 37083210 DOI: 10.1002/adma.202210885] [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: 11/22/2022] [Revised: 01/18/2023] [Indexed: 05/03/2023]
Abstract
Conjugation of aptamers to homogeneous catalysts ("nucleoapzymes"), heterogeneous nanoparticle catalysts ("aptananozymes"), and photocatalysts ("photoaptazymes") yields superior catalytic/photocatalytic hybrid nanostructures emulating functions of native enzymes and photosystems. The concentration of the substrate in proximity to the catalytic sites ("molarity effect") or spatial concentration of electron-acceptor units in spatial proximity to the photosensitizers, by aptamer-ligand complexes, leads to enhanced catalytic/photocatalytic efficacies of the hybrid nanostructures. This is exemplified by sets of "nucleoapzymes" composed of aptamers conjugated to the hemin/G-quadruplex DNAzymes or metal-ligand complexes as catalysts, catalyzing the oxidation of dopamine to aminochrome, oxygen-insertion into the Ar─H moiety of tyrosinamide and the subsequent oxidation of the catechol product into aminochrome, or the hydrolysis of esters or ATP. Also, aptananozymes consisting of aptamers conjugated to Cu2+ - or Ce4+ -ion-modified C-dots or polyadenine-stabilized Au nanoparticles acting as catalysts oxidizing dopamine or operating bioreactor biocatalytic cascades, are demonstrated. In addition, aptamers conjugated to the Ru(II)-tris-bipyridine photosensitizer or the Zn(II) protoporphyrin IX photosensitizer provide supramolecular photoaptazyme assemblies emulating native photosynthetic reaction centers. Effective photoinduced electron transfer followed by the catalyzed synthesis of NADPH or the evolution of H2 is demonstrated by the photosystems. Structure-function relationships dictate the catalytic and photocatalytic efficacies of the systems.
Collapse
Affiliation(s)
- Yu Ouyang
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Michael P O'Hagan
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Bilha Willner
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Itamar Willner
- The Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| |
Collapse
|
3
|
Teodori L, Omer M, Kjems J. RNA nanostructures for targeted drug delivery and imaging. RNA Biol 2024; 21:1-19. [PMID: 38555519 PMCID: PMC10984137 DOI: 10.1080/15476286.2024.2328440] [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] [Accepted: 03/04/2024] [Indexed: 04/02/2024] Open
Abstract
The RNA molecule plays a pivotal role in many biological processes by relaying genetic information, regulating gene expression, and serving as molecular machines and catalyzers. This inherent versatility of RNA has fueled significant advancements in the field of RNA nanotechnology, driving the engineering of complex nanoscale architectures toward biomedical applications, including targeted drug delivery and bioimaging. RNA polymers, serving as building blocks, offer programmability and predictability of Watson-Crick base pairing, as well as non-canonical base pairing, for the construction of nanostructures with high precision and stoichiometry. Leveraging the ease of chemical modifications to protect the RNA from degradation, researchers have developed highly functional and biocompatible RNA architectures and integrated them into preclinical studies for the delivery of payloads and imaging agents. This review offers an educational introduction to the use of RNA as a biopolymer in the design of multifunctional nanostructures applied to targeted delivery in vivo, summarizing physical and biological barriers along with strategies to overcome them. Furthermore, we highlight the most recent progress in the development of both small and larger RNA nanostructures, with a particular focus on imaging reagents and targeted cancer therapeutics in pre-clinical models and provide insights into the prospects of this rapidly evolving field.
Collapse
Affiliation(s)
- Laura Teodori
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus, Denmark
- Center for RNA Therapeutics towards Metabolic Diseases (RNA-META), Aarhus University, Aarhus, Denmark
| | - Marjan Omer
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus, Denmark
- Center for RNA Therapeutics towards Metabolic Diseases (RNA-META), Aarhus University, Aarhus, Denmark
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
4
|
Sequeira-Antunes B, Ferreira HA. Nucleic Acid Aptamer-Based Biosensors: A Review. Biomedicines 2023; 11:3201. [PMID: 38137422 PMCID: PMC10741014 DOI: 10.3390/biomedicines11123201] [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: 10/20/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Aptamers, short strands of either DNA, RNA, or peptides, known for their exceptional specificity and high binding affinity to target molecules, are providing significant advancements in the field of health. When seamlessly integrated into biosensor platforms, aptamers give rise to aptasensors, unlocking a new dimension in point-of-care diagnostics with rapid response times and remarkable versatility. As such, this review aims to present an overview of the distinct advantages conferred by aptamers over traditional antibodies as the molecular recognition element in biosensors. Additionally, it delves into the realm of specific aptamers made for the detection of biomarkers associated with infectious diseases, cancer, cardiovascular diseases, and metabolomic and neurological disorders. The review further elucidates the varying binding assays and transducer techniques that support the development of aptasensors. Ultimately, this review discusses the current state of point-of-care diagnostics facilitated by aptasensors and underscores the immense potential of these technologies in advancing the landscape of healthcare delivery.
Collapse
Affiliation(s)
- Beatriz Sequeira-Antunes
- Institute of Biophysics and Biomedical Engineering, Faculty of Sciences, University of Lisbon, Campo Grande, 1749-016 Lisboa, Portugal
- Exotictarget, 4900-378 Viana do Castelo, Portugal
- Instituto de Engenharia de Sistemas e Computadores-Microsistemas e Nanotecnologias (INESC-MN), 1000-029 Lisbon, Portugal
| | - Hugo Alexandre Ferreira
- Institute of Biophysics and Biomedical Engineering, Faculty of Sciences, University of Lisbon, Campo Grande, 1749-016 Lisboa, Portugal
- Exotictarget, 4900-378 Viana do Castelo, Portugal
| |
Collapse
|
5
|
Investigating Efficacy of Three DNA-Aptamers in Targeted Plasmid Delivery to Human Prostate Cancer Cell Lines. Mol Biotechnol 2023; 65:97-107. [PMID: 35834121 DOI: 10.1007/s12033-022-00528-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/23/2022] [Indexed: 01/11/2023]
Abstract
Selection of targeted and efficient carriers to deliver drugs and genes to cells and tissues is still a major challenge and to overcome this obstacle, aptamers conjugated to nanoparticles have been broadly examined. To assess whether polycation of aptamers can improve plasmid delivery efficacy, we investigated the effect of three DNA-aptamers (AS1411, WY-5a, and Sgs-8) conjugated to branched polyethylenimine (b-PEI; MW ∼25 kDa) with different combinations of gene (plasmid) for delivery to prostate cancer cell lines (DU145 and PC3). According to transfection assessments, the dual conjugation of aptamers (AS:WY) with b-PEI produced the best results and increased the efficiency of plasmid delivery to up to three folds compared to unmodified PEI. Surprisingly, triple aptamer arrangement not only reduced transfection ability but also showed cytotoxicity. While our results demonstrated potential synergistic effects of AS1411 and WY-5a aptamers for gene delivery, it is important to note that the present evidence relies on the aptamer and cell types.
Collapse
|
6
|
Ouyang Y, Fadeev M, Zhang P, Carmieli R, Sohn YS, Karmi O, Qin Y, Chen X, Nechushtai R, Willner I. Aptamer-Functionalized Ce 4+-Ion-Modified C-Dots: Peroxidase Mimicking Aptananozymes for the Oxidation of Dopamine and Cytotoxic Effects toward Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2022; 14:55365-55375. [PMID: 36475576 PMCID: PMC9782376 DOI: 10.1021/acsami.2c16199] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Abstract
Aptamer-functionalized Ce4+-ion-modified C-dots act as catalytic hybrid systems, aptananozymes, catalyzing the H2O2 oxidation of dopamine. A series of aptananozymes functionalized with different configurations of the dopamine binding aptamer, DBA, are introduced. All aptananozymes reveal substantially enhanced catalytic activities as compared to the separated Ce4+-ion-modified C-dots and aptamer constituents, and structure-catalytic functions between the structure and binding modes of the aptamers linked to the C-dots are demonstrated. The enhanced catalytic functions of the aptananozymes are attributed to the aptamer-induced concentration of the reaction substrates in spatial proximity to the Ce4+-ion-modified C-dots catalytic sites. The oxidation processes driven by the Ce4+-ion-modified C-dots involve the formation of reactive oxygen species (•OH radicals). Accordingly, Ce4+-ion-modified C-dots with the AS1411 aptamer or MUC1 aptamer, recognizing specific biomarkers associated with cancer cells, are employed as targeted catalytic agents for chemodynamic treatment of cancer cells. Treatment of MDA-MB-231 breast cancer cells and MCF-10A epithelial breast cells, as control, with the AS1411 aptamer- or MUC1 aptamer-modified Ce4+-ion-modified C-dots reveals selective cytotoxicity toward the cancer cells. In vivo experiments reveal that the aptamer-functionalized nanoparticles inhibit MDA-MB-231 tumor growth.
Collapse
Affiliation(s)
- Yu Ouyang
- The
Institute of Chemistry, The Hebrew University
of Jerusalem, Jerusalem 91904, Israel
| | - Michael Fadeev
- The
Institute of Chemistry, The Hebrew University
of Jerusalem, Jerusalem 91904, Israel
| | - Pu Zhang
- The
Institute of Chemistry, The Hebrew University
of Jerusalem, Jerusalem 91904, Israel
| | - Raanan Carmieli
- Department
of Chemical Research Support, Weizmann Institute
of Science, Rehovot 76100, Israel
| | - Yang Sung Sohn
- Institute
of Life Science, The Hebrew University of
Jerusalem, Jerusalem 91904, Israel
| | - Ola Karmi
- Institute
of Life Science, The Hebrew University of
Jerusalem, Jerusalem 91904, Israel
| | - Yunlong Qin
- The
Institute of Chemistry, The Hebrew University
of Jerusalem, Jerusalem 91904, Israel
| | - Xinghua Chen
- The
Institute of Chemistry, The Hebrew University
of Jerusalem, Jerusalem 91904, Israel
| | - Rachel Nechushtai
- Institute
of Life Science, The Hebrew University of
Jerusalem, Jerusalem 91904, Israel
| | - Itamar Willner
- The
Institute of Chemistry, The Hebrew University
of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
7
|
Ouyang Y, Fadeev M, Zhang P, Carmieli R, Li J, Sohn YS, Karmi O, Nechushtai R, Pikarsky E, Fan C, Willner I. Aptamer-Modified Au Nanoparticles: Functional Nanozyme Bioreactors for Cascaded Catalysis and Catalysts for Chemodynamic Treatment of Cancer Cells. ACS NANO 2022; 16:18232-18243. [PMID: 36286233 PMCID: PMC9706657 DOI: 10.1021/acsnano.2c05710] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Polyadenine-stabilized Au nanoparticles (pA-AuNPs) reveal dual nanozyme catalytic activities toward the H2O2-mediated oxidation of dopamine to aminochrome and toward the aerobic oxidation of glucose to gluconic acid and H2O2. The conjugation of a dopamine-binding aptamer (DBA) to the pA-AuNPs yields aptananozyme structures catalyzing simultaneously the H2O2-mediated oxidation of dopamine to aminochrome through the aerobic oxidation of glucose. A set of aptananozymes consisting of DBA conjugated through the 5'- or 3'-end directly or spacer bridges to pA-AuNPs were synthesized. The set of aptananozymes revealed enhanced catalytic activities toward the H2O2-catalyzed oxidation of dopamine to dopachrome, as compared to the separated pA-AuNPs and DBA constituents, and structure-function relationships within the series of aptananozymes were demonstrated. The enhanced catalytic function of the aptananozymes was attributed to the concentration of the dopamine at the catalytic interfaces by means of aptamer-dopamine complexes. The dual catalytic activities of aptananozymes were further applied to design bioreactors catalyzing the effective aerobic oxidation of dopamine in the presence of glucose. Mechanistic studies demonstrated that the aptananozymes generate reactive oxygen species. Accordingly, the AS1411 aptamer, recognizing the nucleolin receptor associated with cancer cells, was conjugated to the pA-AuNPs, yielding a nanozyme for the chemodynamic treatment of cancer cells. The AS1411 aptamer targets the aptananozyme to the cancer cells and facilitates the selective permeation of the nanozyme into the cells. Selective cytotoxicity toward MDA-MB-231 breast cancer cells (ca. 70% cell death) as compared to MCF-10A epithelial cells (ca. 2% cell death) is demonstrated.
Collapse
Affiliation(s)
- Yu Ouyang
- The
Institute of Chemistry, The Hebrew University
of Jerusalem, Jerusalem 91904, Israel
| | - Michael Fadeev
- The
Institute of Chemistry, The Hebrew University
of Jerusalem, Jerusalem 91904, Israel
| | - Pu Zhang
- The
Institute of Chemistry, The Hebrew University
of Jerusalem, Jerusalem 91904, Israel
| | - Raanan Carmieli
- Department
of Chemical Research Support, Weizmann Institute
of Science, Rehovot 76100, Israel
| | - Jiang Li
- School
of Chemistry and Chemical Engineering, Frontiers Science Center for
Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- The
Interdisciplinary Research Center, Shanghai Synchrotron Radiation
Facility, Zhangjiang Laboratory, Shanghai
Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yang Sung Sohn
- Institute
of Life Science, The Hebrew University of
Jerusalem, Jerusalem 91904, Israel
| | - Ola Karmi
- Institute
of Life Science, The Hebrew University of
Jerusalem, Jerusalem 91904, Israel
| | - Rachel Nechushtai
- Institute
of Life Science, The Hebrew University of
Jerusalem, Jerusalem 91904, Israel
| | - Eli Pikarsky
- The Lautenberg
Center for Immunology and Cancer Research, IMRIC, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Chunhai Fan
- School
of Chemistry and Chemical Engineering, Frontiers Science Center for
Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Itamar Willner
- The
Institute of Chemistry, The Hebrew University
of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
8
|
Hu X, Zhang D, Zeng Z, Huang L, Lin X, Hong S. Aptamer-Based Probes for Cancer Diagnostics and Treatment. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111937. [PMID: 36431072 PMCID: PMC9695321 DOI: 10.3390/life12111937] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/23/2022] [Accepted: 11/12/2022] [Indexed: 11/22/2022]
Abstract
Aptamers are single-stranded DNA or RNA oligomers that have the ability to generate unique and diverse tertiary structures that bind to cognate molecules with high specificity. In recent years, aptamer researches have witnessed a huge surge, owing to its unique properties, such as high specificity and binding affinity, low immunogenicity and toxicity, and simplicity of synthesis with negligible batch-to-batch variation. Aptamers may bind to targets, such as various cancer biomarkers, making them applicable for a wide range of cancer diagnosis and treatment. In cancer diagnostic applications, aptamers are used as molecular probes instead of antibodies. They have the potential to detect various cancer-associated biomarkers. For cancer therapeutic purposes, aptamers can serve as therapeutic or delivery agents. The chemical stabilization and modification strategies for aptamers may expand their serum half-life and shelf life. However, aptamer-based probes for cancer diagnosis and therapy still face several challenges for successful clinical translation. A deeper understanding of nucleic acid chemistry, tissue distribution, and pharmacokinetics is required in the development of aptamer-based probes. This review summarizes their application in cancer diagnostics and treatments based on different localization of target biomarkers, as well as current challenges and future prospects.
Collapse
|
9
|
Technetium-99m radiolabeled nucleolin-targeted aptamer for glioma tumor imaging in murine models. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.130587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
10
|
Cruz-Hernández CD, Rodríguez-Martínez G, Cortés-Ramírez SA, Morales-Pacheco M, Cruz-Burgos M, Losada-García A, Reyes-Grajeda JP, González-Ramírez I, González-Covarrubias V, Camacho-Arroyo I, Cerbón M, Rodríguez-Dorantes M. Aptamers as Theragnostic Tools in Prostate Cancer. Biomolecules 2022; 12:biom12081056. [PMID: 36008950 PMCID: PMC9406110 DOI: 10.3390/biom12081056] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 02/07/2023] Open
Abstract
Despite of the capacity that several drugs have for specific inhibition of the androgen receptor (AR), in most cases, PCa progresses to an androgen-independent stage. In this context, the development of new targeted therapies for prostate cancer (PCa) has remained as a challenge. To overcome this issue, new tools, based on nucleic acids technology, have been developed. Aptamers are small oligonucleotides with a three-dimensional structure capable of interacting with practically any desired target, even large targets such as mammalian cells or viruses. Recently, aptamers have been studied for treatment and detection of many diseases including cancer. In PCa, numerous works have reported their use in the development of new approaches in diagnostics and treatment strategies. Aptamers have been joined with drugs or other specific molecules such as silencing RNAs (aptamer–siRNA chimeras) to specifically reduce the expression of oncogenes in PCa cells. Even though these studies have shown good results in the early stages, more research is still needed to demonstrate the clinical value of aptamers in PCa. The aim of this review was to compile the existing scientific literature regarding the use of aptamers in PCa in both diagnosis and treatment studies. Since Prostate-Specific Membrane Antigen (PSMA) aptamers are the most studied type of aptamers in this field, special emphasis was given to these aptamers.
Collapse
Affiliation(s)
- Carlos David Cruz-Hernández
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
| | - Griselda Rodríguez-Martínez
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
| | - Sergio A. Cortés-Ramírez
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
| | - Miguel Morales-Pacheco
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
| | - Marian Cruz-Burgos
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
| | - Alberto Losada-García
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
| | - Juan Pablo Reyes-Grajeda
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico;
| | - Imelda González-Ramírez
- Departamento de Atención a la Salud, Universidad Autónoma Metropolitana–Xochimilco, Mexico City 04960, Mexico;
| | | | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico; (I.C.-A.); (M.C.)
| | - Marco Cerbón
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico; (I.C.-A.); (M.C.)
| | - Mauricio Rodríguez-Dorantes
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico; (C.D.C.-H.); (G.R.-M.); (S.A.C.-R.); (M.M.-P.); (M.C.-B.); (A.L.-G.)
- Correspondence:
| |
Collapse
|
11
|
Kozani PS, Kozani PS, Malik MT. AS1411-functionalized delivery nanosystems for targeted cancer therapy. EXPLORATION OF MEDICINE 2021; 2:146-166. [PMID: 34723284 PMCID: PMC8555908 DOI: 10.37349/emed.2021.00039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/27/2021] [Indexed: 12/12/2022] Open
Abstract
Nucleolin (NCL) is a multifunctional nucleolar phosphoprotein harboring critical roles in cells such as cell proliferation, survival, and growth. The dysregulation and overexpression of NCL are related to various pathologic and oncological indications. These characteristics of NCL make it an ideal target for the treatment of various cancers. AS1411 is a synthetic quadruplex-forming nuclease-resistant DNA oligonucleotide aptamer which shows a considerably high affinity for NCL, therefore, being capable of inducing growth inhibition in a variety of tumor cells. The high affinity and specificity of AS1411 towards NCL make it a suitable targeting tool, which can be used for the functionalization of therapeutic payloaddelivery nanosystems to selectively target tumor cells. This review explores the advances in NCL-targeting cancer therapy through AS1411-functionalized delivery nanosystems for the selective delivery of a broad spectrum of therapeutic agents.
Collapse
Affiliation(s)
- Pooria Safarzadeh Kozani
- Carlos Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14115/111, Iran
| | - Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht 41446/66949, Iran
- Student Research Committee, Medical Biotechnology Research Center, School of Nursing, Midwifery, and Paramedicine, Guilan University of Medical Sciences, Rasht 41446/66949, Iran
| | - Mohammad Tariq Malik
- Departments of Microbiology and Immunology, Regenerative Medicine, and Stem Cell Biology, University of Louisville, Louisville, KY 40202, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
12
|
Piramoon M, Khodadust F, Hosseinimehr SJ. Radiolabeled nanobodies for tumor targeting: From bioengineering to imaging and therapy. Biochim Biophys Acta Rev Cancer 2021; 1875:188529. [PMID: 33647388 DOI: 10.1016/j.bbcan.2021.188529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 02/08/2023]
Abstract
So far, numerous molecules and biomolecules have been evaluated for tumor targeting purposes for radionuclide-based imaging and therapy modalities. Due to the high affinity and specificity against tumor antigens, monoclonal antibodies are appropriate candidates for tumor targeting. However, their large size prevents their comprehensive application in radionuclide-based tumor imaging or therapy, since it leads to their low tumor penetration, low blood clearance, and thus inappropriate tumor-to-background ratio. Nowadays, the variable domain of heavy-chain antibodies from the Camelidae family, known as nanobodies (Nbs), turn into exciting candidates for medical research. Considering several innate advantages of these new tumor-targeting agents, including excellent affinity and specificity toward antigen, high solubility, high stability, fast washout from blood, convenient production, ease of selection, and low immunogenicity, it assumes that they may overcome generic problems of monoclonal antibodies, their fragments, and other vectors used for tumor imaging/therapy. After three decades of Nbs discovery, the increasing number of their preclinical and clinical investigations, which have led to outstanding results, confirm their application for tumor targeting purposes. This review describes Nbs characteristics, the diagnostic and therapeutic application of their radioconjugates, and their recent advances.
Collapse
Affiliation(s)
- Majid Piramoon
- Department of Medicinal Chemistry and Radiopharmacy, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran; Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Fatemeh Khodadust
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
13
|
Sheetz T, Mills J, Tessari A, Pawlikowski M, Braddom AE, Posid T, Zynger DL, James C, Embrione V, Parbhoo K, Foray C, Coppola V, Croce CM, Palmieri D. NCL Inhibition Exerts Antineoplastic Effects against Prostate Cancer Cells by Modulating Oncogenic MicroRNAs. Cancers (Basel) 2020; 12:E1861. [PMID: 32664322 PMCID: PMC7408652 DOI: 10.3390/cancers12071861] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/05/2020] [Accepted: 07/06/2020] [Indexed: 11/16/2022] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer in men and second most common cause of cancer-related deaths in the United States. Androgen deprivation therapy (ADT) is only temporarily effective for advanced-stage PCa, as the disease inevitably progresses to castration-resistant prostate cancer (CRPC). The protein nucleolin (NCL) is overexpressed in several types of human tumors where it is also mislocalized to the cell surface. We previously reported the identification of a single-chain fragment variable (scFv) immuno-agent that is able to bind NCL on the surface of breast cancer cells and inhibit proliferation both in vitro and in vivo. In the present study, we evaluated whether NCL could be a valid therapeutic target for PCa, utilizing DU145, PC3 (CRPC), and LNCaP (androgen-sensitive) cell lines. First, we interrogated the publicly available databases and noted that higher NCL mRNA levels are associated with higher Gleason Scores as well as with recurrent and metastatic tumors. Then, using our anti-NCL scFv, we demonstrated that NCL is expressed on the surface of all three tested cell lines and that NCL inhibition results in reduced proliferation and migration. We also measured the inhibitory effect of NCL targeting on the biogenesis of oncogenic microRNAs such as miR-21, -221 and -222, which was cell context dependent. Taken together, our data provide evidence that NCL targeting inhibits the key hallmarks of malignancy in PCa cells and may provide a novel therapeutic option for patients with advanced-stage PCa.
Collapse
Affiliation(s)
- Tyler Sheetz
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Department of Urology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Joseph Mills
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Anna Tessari
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Megan Pawlikowski
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Ashley E. Braddom
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Tasha Posid
- Department of Urology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Debra L. Zynger
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Cindy James
- Mass Spectroscopy and Proteomics Facility, The Ohio State University, Columbus, OH 43210, USA;
| | - Valerio Embrione
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Kareesma Parbhoo
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Claudia Foray
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Carlo M. Croce
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Dario Palmieri
- Department of Cancer Biology and Genetics, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (T.S.); (J.M.); (A.T.); (M.P.); (A.E.B.); (V.E.); (K.P.); (C.F.); (V.C.); (C.M.C.)
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
14
|
Zhang GX, Liu YL, Yang M, Huang WS, Xu JH. An aptamer-based, fluorescent and radionuclide dual-modality probe. Biochimie 2020; 171-172:55-62. [PMID: 32081705 DOI: 10.1016/j.biochi.2020.02.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 02/13/2020] [Indexed: 12/25/2022]
Abstract
Aptamers which are promising and effective molecular probes, can deliver either fluorescent materials or radionuclides to tumors. This study aimed to develop a novel both fluorescent and radionuclide dual-modality probe based on a truncated aptamer and evaluate its stability and binding affinities in vitro. The aptamer JHIT2 with binding specifically to HepG2 cells was previously generated by Cell-SELEX. Using mfold and RNAstructure software to predict the secondary structure folded by a middle random sequence to truncate the primer sequences at both ends of the aptamer JHIT2 to yield the aptamer JHIT2e, with a similar secondary structure to JHIT2 and the same specificity and affinity as JHIT2. Attaching carboxyfluorescein (FAM) readily to the aptamer JHIT2e and then attaching iodine-131 to the FAM moiety which has multiple sites for iodine labeling to develop a novel both fluorescent and radionuclide dual-modality probe, termed 131I-FAM-JHIT2e. Cell uptake and fluorescence imaging assays in vitro confirmed that 131I-FAM-JHIT2e had both FAM fluorescence signal and radio-activity signal and maintained specific binding ability to the human hepatoma cell line HepG2. This work formed a basis for aptamer-based, dual-modality imaging probe that contains both fluorescent and radionuclide tags, which also is potential for theranostics.
Collapse
Affiliation(s)
- Gui-Xiong Zhang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China.
| | - Yan-Lan Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, 410082, China.
| | - Min Yang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Wen-Shan Huang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China
| | - Jie-Hua Xu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, China.
| |
Collapse
|
15
|
Li H, Liu J, Xiao X, Sun S, Zhang H, Zhang Y, Zhou W, Zhang B, Roy M, Liu H, Ye M, Wang Z, Liu-Smith F, Liu J. A Novel Aptamer LL4A Specifically Targets Vemurafenib-Resistant Melanoma through Binding to the CD63 Protein. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:727-738. [PMID: 31726389 PMCID: PMC6859286 DOI: 10.1016/j.omtn.2019.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 09/29/2019] [Accepted: 10/08/2019] [Indexed: 01/08/2023]
Abstract
Melanoma is a highly aggressive tumor with a poor prognosis, and half of all melanoma patients harbor BRAF mutations. A BRAF inhibitor, vemurafenib (PLX4032), has been approved by the US Food and Drug Administration (FDA) and European Medicines Agency (EMA) to treat advanced melanoma patients with BRAFV600E mutation. However, the efficacy of vemurafenib is impeded by adaptive resistance in almost all patients. In this study, using a cell-based SELEX (systematic evolution of ligands by exponential enrichment) strategy, we obtained a DNA aptamer (named LL4) with high affinity and specificity against vemurafenib-resistant melanoma cells. Optimized truncated form (LL4A) specifically binds to vemurafenib-resistant melanoma cells with dissociation constants in the nanomolar range and with excellent stability and low toxicity. Meanwhile, fluorescence imaging confirmed that LL4A significantly accumulated in tumors formed by vemurafenib-resistant melanoma cells, but not in control tumors formed by their corresponding parental cells in vivo. Further, a transmembrane protein CD63 was identified as the binding target of aptamer LL4A using a pull-down assay combined with the liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. CD63 formed a supramolecular complex with TIMP1 and β1-integrin, activated the nuclear factor кB (NF-кB) and mitogen-activated protein kinase (MAPK) signaling pathways, and contributed to vemurafenib resistance. Potentially, the aptamer LL4A may be used diagnostically and therapeutically in humans to treat targeted vemurafenib-resistant melanoma.
Collapse
Affiliation(s)
- Hui Li
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China
| | - Juan Liu
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China
| | - Xiaojuan Xiao
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China
| | - Shuming Sun
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China
| | - Hui Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
| | - Yibin Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
| | - Weihua Zhou
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; Department of Obstetrics and Gynecology, People's Hospital of Xiangxi Tujia and Miao Autonomous Prefecture, Hunan, Jishou 410006, China
| | - Bin Zhang
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Mridul Roy
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
| | - Hong Liu
- The First Xiangya Hospital, Central South University, Changsha 410078, China
| | - Mao Ye
- Molecular Science and Biomedicine Laboratory, State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, China
| | - Zi Wang
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; The First Xiangya Hospital, Central South University, Changsha 410078, China.
| | - Feng Liu-Smith
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; Department of Epidemiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA.
| | - Jing Liu
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China.
| |
Collapse
|
16
|
Clinical aspects of radiolabeled aptamers in diagnostic nuclear medicine: A new class of targeted radiopharmaceuticals. Bioorg Med Chem 2018; 27:2282-2291. [PMID: 30502114 DOI: 10.1016/j.bmc.2018.11.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/16/2018] [Accepted: 11/21/2018] [Indexed: 12/31/2022]
Abstract
Targeted radiopharmaceuticals offer the possibility of improved imaging with reduced side effects. Up to now, a variety of biological receptors such as aptamers have been successfully radiolabeled and applied to diagnostic imaging of cancers. The concept of using radio-labeled aptamers for binding to their targets has stimulated an immense body of research in diagnostic nuclear medicine. These biological recognition elements are single-stranded oligonucleotides that interact with their target molecules with high affinity and specificity in unique three-dimensional structures. Because of their high affinity and specificity, the receptor-binding aptamers labeled with gamma emitters such as 99mTc, 64Cu, 111In, 18F and 67Ga can facilitate the visualization of receptor-expressing tissues noninvasively. Compared to the antibody-based radiopharmaceuticals, the radiolabeled aptamers provide a number of advantages for clinical diagnostics including high stability, low cost, and ease of production and modification, low immunogenicity and, especially, superior tissue penetration because of their smaller size. In this review, we present recent progresses and challenges in aptamer-based diagnostic radiopharmaceuticals and highlight some representative applications of aptamers in nuclear medicine.
Collapse
|
17
|
Hassanzadeh L, Chen S, Veedu RN. Radiolabeling of Nucleic Acid Aptamers for Highly Sensitive Disease-Specific Molecular Imaging. Pharmaceuticals (Basel) 2018; 11:E106. [PMID: 30326601 PMCID: PMC6315947 DOI: 10.3390/ph11040106] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/06/2018] [Accepted: 10/10/2018] [Indexed: 12/14/2022] Open
Abstract
Aptamers are short single-stranded DNA or RNA oligonucleotide ligand molecules with a unique three-dimensional shape, capable of binding to a defined molecular target with high affinity and specificity. Since their discovery, aptamers have been developed for various applications, including molecular imaging, particularly nuclear imaging that holds the highest potential for the clinical translation of aptamer-based molecular imaging probes. Their easy laboratory production without any batch-to-batch variations, their high stability, their small size with no immunogenicity and toxicity, and their flexibility to incorporate various functionalities without compromising the target binding affinity and specificity make aptamers an attractive class of targeted-imaging agents. Aptamer technology has been utilized in nuclear medicine imaging techniques, such as single photon emission computed tomography (SPECT) and positron emission tomography (PET), as highly sensitive and accurate biomedical imaging modalities towards clinical diagnostic applications. However, for aptamer-targeted PET and SPECT imaging, conjugation of appropriate radionuclides to aptamers is crucial. This review summarizes various strategies to link the radionuclides to chemically modified aptamers to accomplish aptamer-targeted PET and SPECT imaging.
Collapse
Affiliation(s)
- Leila Hassanzadeh
- Department of Nuclear Medicine, School of Medicine, Rajaie Cardiovascular, Medical and Research Center & Department of Medicinal Chemistry, School of Pharmacy-International Campus, Iran University of Medical Sciences, Tehran 1449614535, Iran.
- Centre for Comparative Genomics, Murdoch University, Perth 6150, Australia.
| | - Suxiang Chen
- Centre for Comparative Genomics, Murdoch University, Perth 6150, Australia.
- Perron Institute for Neurological and Translational Science, Perth 6009, Australia.
| | - Rakesh N Veedu
- Centre for Comparative Genomics, Murdoch University, Perth 6150, Australia.
- Perron Institute for Neurological and Translational Science, Perth 6009, Australia.
| |
Collapse
|
18
|
Molavipordanjani S, Tolmachev V, Hosseinimehr SJ. Basic and practical concepts of radiopharmaceutical purification methods. Drug Discov Today 2018; 24:315-324. [PMID: 30278224 DOI: 10.1016/j.drudis.2018.09.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/16/2018] [Accepted: 09/26/2018] [Indexed: 01/02/2023]
Abstract
The presence of radiochemical impurities in a radiopharmaceutical contributes to an unnecessary radiation burden for the patients or to an undesirable high radioactivity background, which reduces the imaging contrast or therapeutic efficacy. Therefore, if the radiolabeling process results in unsatisfactory radiochemical purity, a purification step is unavoidable. A successful purification process requires a profound knowledge about the radiopharmaceuticals of interest ranging from structural features to susceptibility to different conditions. Most radiopharmaceutical purification methods are based on solid-phase extraction (SPE), high-performance liquid chromatography (HPLC), size exclusion chromatography (SEC), ion-exchange chromatography (IEC), and liquid-liquid extraction (LLE). Here, we discuss the basic and applied concepts of these purifications methods as well as their advantages and limitations.
Collapse
Affiliation(s)
- Sajjad Molavipordanjani
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
19
|
Application of aptamers for in vivo molecular imaging and theranostics. Adv Drug Deliv Rev 2018; 134:94-106. [PMID: 30125606 DOI: 10.1016/j.addr.2018.08.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/06/2018] [Accepted: 08/16/2018] [Indexed: 12/11/2022]
Abstract
Nucleic acid aptamers are small three-dimensional structures of oligonucleotides selected to bind to a target of interest with high affinity and specificity. In vitro, aptamers already compete with antibodies to serve as imaging probes, e.g. for microscopy or flow cytometry. However, they are also increasingly used for in vivo molecular imaging. Accordingly, aptamers have been evaluated over the last twenty years in almost every imaging modality, including single photon emission computed tomography, positron emission tomography, magnetic resonance imaging, fluorescence imaging, echography, and x-ray computed tomography. This review focuses on the studies that were conducted in vivo with aptamer-based imaging probes. It also presents how aptamers have been recently used to develop new types of probes for multimodal imaging and theranostic applications.
Collapse
|
20
|
Riccardi C, Fàbrega C, Grijalvo S, Vitiello G, D'Errico G, Eritja R, Montesarchio D. AS1411-decorated niosomes as effective nanocarriers for Ru(iii)-based drugs in anticancer strategies. J Mater Chem B 2018; 6:5368-5384. [PMID: 32254501 DOI: 10.1039/c8tb01563e] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Niosomes are self-assembled vesicles made up of single chain non-ionic surfactants combined with appropriate amounts of cholesterol or other lipids, exploited as carriers for hydrophilic or lipophilic drugs. Compared to liposomes, niosomes are typically more stable, less expensive and, being generally obtained from synthetic surfactants, more easily derivatizable, providing vesicular structures with a higher versatility and chemical diversity. Herein, we investigated the physico-chemical and biological properties of niosomes loaded with two active ingredients, i.e. the nucleolipidic Ru(iii)-complex HoThyRu, selected as an anticancer agent, and the nucleolin-targeting AS1411 aptamer, allowing selective recognition of cancer cells. The morphology, average size, zeta potential, electrophoretic mobility, and stability over time of the functionalized niosomes were analyzed using different biophysical techniques. These formulations, tested on both cancer and normal cells, showed promising antiproliferative activity on HeLa cells, with a higher efficacy associated with the nanosystems containing both AS1411 and HoThyRu with respect to the controls. In all the tested cell lines, AS1411 proved to markedly enhance the bioactivity of the Ru(iii)-containing niosomes.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126, Napoli, Italy.
| | | | | | | | | | | | | |
Collapse
|
21
|
Riccardi C, Musumeci D, Russo Krauss I, Piccolo M, Irace C, Paduano L, Montesarchio D. Exploring the conformational behaviour and aggregation properties of lipid-conjugated AS1411 aptamers. Int J Biol Macromol 2018; 118:1384-1399. [PMID: 30170359 DOI: 10.1016/j.ijbiomac.2018.06.137] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 06/23/2018] [Accepted: 06/26/2018] [Indexed: 12/19/2022]
Abstract
AS1411 is a nucleolin-binding aptamer which attracted great interest as active targeting ligand for the selective delivery of therapeutic agents to tumour cells. In this work we selected three AS1411 derivatives 5'-conjugated with lipophilic tails and studied their properties in view of their application in liposomial formulations and/or lipid coated-nanoparticles for targeted therapies. The conformational behaviour of these AS1411 analogs has been investigated in comparison with the unmodified aptamer by CD, UV, PAGE, SEC-HPLC, DLS and thioflavin T (ThT) fluorescence assays to get insight in their secondary structure and aggregation properties. This study has been performed in pseudo-physiological buffers mimicking the extra- and intracellular environments, and at different concentrations in the μM range, paying special attention to the effects of the lipophilic tail on the overall aptamer conformation. The 5'-lipidated AS1411 derivatives proved to fold into stable, parallel unimolecular G-quadruplex structures, forming large aggregates, mainly micelles, at conc. >10 μM. Preliminary bioscreenings on selected cancer cells showed that these derivatives are less cytotoxic than AS1411, but maintain a similar biological behaviour. This study demonstrated that lipophilic tails dramatically favour the formation of AS1411 aggregates, however not impairing the formation and thermal stability of its peculiar G4 motifs.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Napoli, Italy
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Napoli, Italy; Institute of Biostructures and Bioimages, CNR, Via Mezzocannone 16, I-80134 Napoli, Italy
| | - Irene Russo Krauss
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Napoli, Italy; CSGI - Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, Via della Lastruccia 3, I-50019 Sesto Fiorentino (Fi), Italy
| | - Marialuisa Piccolo
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
| | - Carlo Irace
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
| | - Luigi Paduano
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Napoli, Italy; CSGI - Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande Interfase, Via della Lastruccia 3, I-50019 Sesto Fiorentino (Fi), Italy
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 21, I-80126 Napoli, Italy; Institute for Endocrinology and Oncology "Gaetano Salvatore", CNR, Via Pansini 5, 80131 Napoli, Italy.
| |
Collapse
|
22
|
Hori SI, Herrera A, Rossi JJ, Zhou J. Current Advances in Aptamers for Cancer Diagnosis and Therapy. Cancers (Basel) 2018; 10:cancers10010009. [PMID: 29301363 PMCID: PMC5789359 DOI: 10.3390/cancers10010009] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 12/22/2017] [Accepted: 12/26/2017] [Indexed: 12/24/2022] Open
Abstract
Nucleic acid aptamers are single-stranded oligonucleotides that interact with target molecules with high affinity and specificity in unique three-dimensional structures. Aptamers are generally isolated by a simple selection process called systematic evolution of ligands by exponential enrichment (SELEX) and then can be chemically synthesized and modified. Because of their high affinity and specificity, aptamers are promising agents for biomarker discovery, as well as cancer diagnosis and therapy. In this review, we present recent progress and challenges in aptamer and SELEX technology and highlight some representative applications of aptamers in cancer therapy.
Collapse
Affiliation(s)
- Shin-Ichiro Hori
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010, USA.
- Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka, Osaka 561-0825, Japan.
| | - Alberto Herrera
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010, USA.
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010, USA.
| | - John J Rossi
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010, USA.
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010, USA.
| | - Jiehua Zhou
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010, USA.
| |
Collapse
|
23
|
Khodadust F, Ahmadpour S, Aligholikhamseh N, Abedi SM, Hosseinimehr SJ. An improved 99mTc-HYNIC-(Ser) 3-LTVSPWY peptide with EDDA/tricine as co-ligands for targeting and imaging of HER2 overexpression tumor. Eur J Med Chem 2017; 144:767-773. [PMID: 29291444 DOI: 10.1016/j.ejmech.2017.12.037] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/10/2017] [Accepted: 12/12/2017] [Indexed: 01/12/2023]
Abstract
Overexpression of human epidermal receptor 2 (HER2) has given the opportunity for targeting and delivering of imaging radiotracers. The aim of this study was to evaluate the 99mTc-HYNIC-(EDDA/tricine)-(Ser)3-LTVSPWY peptide for tumor targeting and imaging of tumor with overexpression of HER2. The HYNIC-(Ser)3-LTVSPWY was labeled with 99mTc in presence of EDDA/tricine mixture as co-ligands. The in vitro and in vivo studies of this radiolabeled peptide were performed for cellular specific binding and tumor targeting. The high radiochemical purity of 99mTc-HYNIC (EDDA/tricine)-(Ser)3-LTVSPWY was obtained to be 99%. It exhibited high stability in normal saline and human serum. In HER2 binding affinity study, a significant reduction in uptake of radiolabeled peptide (7.7 fold) was observed by blocking SKOV-3 cells receptors with unlabeled peptide. The KD and Bmax values for this radiolabeled peptide were determined as 3.3 ± 1.0 nM and 2.9 ± 0.3 × 106 CPM/pMol, respectively. Biodistribution study revealed tumor to blood and tumor to muscle ratios about 6.9 and 4 respectively after 4 h. Tumor imaging by gamma camera demonstrated considerable high contrast tumor uptake. This developed 99mTc-HYNIC-(Ser)3-LTVSPWY peptide selectively targeted on HER2 tumor and exhibited a high target uptake combined with acceptable low background activity for tumor imaging in mice. The results of this study and its comparison with another study showed that 99mTc-HYNIC-(EDDA/tricine)-(Ser)3-LTVSPWY is much better than previously reported radiolabeled peptide as 99mTc-CSSS-LTVSPWY for HER2 overexpression tumor targeting and imaging.
Collapse
Affiliation(s)
- Fatemeh Khodadust
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sajjad Ahmadpour
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nazan Aligholikhamseh
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mohammad Abedi
- Department of Radiology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
24
|
Rahmanian N, Hosseinimehr SJ, Khalaj A, Noaparast Z, Abedi SM, Sabzevari O. 99mTc labeled HYNIC-EDDA/tricine-GE11 peptide as a successful tumor targeting agent. Med Chem Res 2017. [DOI: 10.1007/s00044-017-2111-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
25
|
Hanif S, Liu HL, Ahmed SA, Yang JM, Zhou Y, Pang J, Ji LN, Xia XH, Wang K. Nanopipette-Based SERS Aptasensor for Subcellular Localization of Cancer Biomarker in Single Cells. Anal Chem 2017; 89:9911-9917. [PMID: 28825473 DOI: 10.1021/acs.analchem.7b02147] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Single cell analysis is essential for understanding the heterogeneity, behaviors of cells, and diversity of target analyte in different subcellular regions. Nucleolin (NCL) is a multifunctional protein that is markedly overexpressed in most of the cancer cells. The variant expression levels of NCL in subcellular regions have a marked influence on cancer proliferation and treatments. However, the specificity of available methods to identify the cancer biomarkers is limited because of the high level of subcellular matrix effect. Herein, we proposed a novel technique to increase both the molecular and spectral specificity of cancer diagnosis by using aptamers affinity based portable nanopipette with distinctive surface-enhanced Raman scattering (SERS) activities. The aptamers-functionalized gold-coated nanopipette was used to capture target, while p-mercaptobenzonitrile (MBN) and complementary DNA modified Ag nanoparticles (AgNPs) worked as Raman reporter to produce SERS signal. The SERS signal of Raman nanotag was lost upon NCL capturing via modified DNA aptamers on nanoprobe, which further helped to verify the specificity of nanoprobe. For proof of concept, NCL protein was specifically extracted from different cell lines by aptamers modified SERS active nanoprobe. The nanoprobes manifested specifically good affinity for NCL with a dissociation constant Kd of 36 nM and provided a 1000-fold higher specificity against other competing proteins. Furthermore, the Raman reporter moiety has a vibrational frequency in the spectroscopically silent region (1800-2300 cm-1) with a negligible matrix effect from cell analysis. The subcellular localization and spatial distribution of NCL were successfully achieved in various types of cells, including MCF-7A, HeLa, and MCF-10A cells. This type of probing technique for single cell analysis could lead to the development of a new perspective in cancer diagnosis and treatment at the cellular level.
Collapse
Affiliation(s)
- Sumaira Hanif
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, and ‡State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University , Nanjing 210023, China
| | - Hai-Ling Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, and ‡State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University , Nanjing 210023, China
| | - Saud Asif Ahmed
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, and ‡State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University , Nanjing 210023, China
| | - Jin-Mei Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, and ‡State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University , Nanjing 210023, China
| | - Yue Zhou
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, and ‡State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University , Nanjing 210023, China
| | - Jie Pang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, and ‡State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University , Nanjing 210023, China
| | - Li-Na Ji
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, and ‡State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University , Nanjing 210023, China
| | - Xing-Hua Xia
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, and ‡State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University , Nanjing 210023, China
| | - Kang Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, and ‡State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University , Nanjing 210023, China
| |
Collapse
|
26
|
99mTc-radiolabeled GE11-modified peptide for ovarian tumor targeting. ACTA ACUST UNITED AC 2017; 25:13. [PMID: 28464952 PMCID: PMC5414288 DOI: 10.1186/s40199-017-0179-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 04/25/2017] [Indexed: 01/01/2023]
Abstract
Background Ovarian cancer is a serious threat for women health and the early diagnosis of this cancer might improves the survival rate of patients. The use of the targeted radiopharmaceuticals could be a non-invasive and logical method for tumor imaging. The aim of this study was to radiolabel GE11 peptide as a new specific probe for imaging of ovarian tumor. Methods HYNIC-SSS-GE11 peptide was labeled with 99mTc using tricine as a coligand. The 99mTc-tricine-HYNIC-SSS-GE11 peptide was evaluated for specific cellular binding in three cell lines with different levels of EGFR expression. Tumor targeting was assessed in SKOV3 tumor bearing mice. Results By using tricine as a coligand, labeling yield was more than 98% and the stability of the radiolabelled peptide in human serum up to 4 h was 96%. The in vitro cell uptake test showed that this radiolabeled peptide had a good affinity to SKOV3 cells with dissociation constant of 73 nM. The in vivo results showed a tumor/muscle ratio of 3.2 at 4 h following injection of 99mTc-tricine-HYNIC-SSS-GE11 peptide. Conclusions Results of this study showed that 99mTc-tricine-HYNIC-SSS-GE11 peptide could be a promising tool for diagnosis and staging of ovarian cancer. Graphical Abstract 99mTc-tricine-HYNIC-SSS-GE11, a novl targeted agent for ovarian tumor imaging![]()
Collapse
|
27
|
Bates PJ, Reyes-Reyes EM, Malik MT, Murphy EM, O'Toole MG, Trent JO. G-quadruplex oligonucleotide AS1411 as a cancer-targeting agent: Uses and mechanisms. Biochim Biophys Acta Gen Subj 2017; 1861:1414-1428. [PMID: 28007579 DOI: 10.1016/j.bbagen.2016.12.015] [Citation(s) in RCA: 201] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/16/2016] [Accepted: 12/17/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND AS1411 is a 26-mer G-rich DNA oligonucleotide that forms a variety of G-quadruplex structures. It was identified based on its cancer-selective antiproliferative activity and subsequently determined to be an aptamer to nucleolin, a multifunctional protein that preferentially binds quadruplex nucleic acids and which is present at high levels on the surface of cancer cells. AS1411 has exceptionally efficient cellular internalization compared to non-quadruplex DNA sequences. SCOPE OF REVIEW Recent developments related to AS1411 will be examined, with a focus on its use for targeted delivery of therapeutic and imaging agents. MAJOR CONCLUSIONS Numerous research groups have used AS1411 as a targeting agent to deliver nanoparticles, oligonucleotides, and small molecules into cancer cells. Studies in animal models have demonstrated that AS1411-linked materials can accumulate selectively in tumors following systemic administration. The mechanism underlying the cancer-targeting ability of AS1411 is not completely understood, but recent studies suggest a model that involves: (1) initial uptake by macropinocytosis, a form of endocytosis prevalent in cancer cells; (2) stimulation of macropinocytosis by a nucleolin-dependent mechanism resulting in further uptake; and (3) disruption of nucleolin-mediated trafficking and efflux leading to cargoes becoming trapped inside cancer cells. SIGNIFICANCE Human trials have indicated that AS1411 is safe and can induce durable remissions in a few patients, but new strategies are needed to maximize its clinical impact. A better understanding of the mechanisms by which AS1411 targets and kills cancer cells may hasten the development of promising technologies using AS1411-linked nanoparticles or conjugates for cancer-targeted therapy and imaging. This article is part of a Special Issue entitled "G-quadruplex" Guest Editor: Dr. Concetta Giancola and Dr. Daniela Montesarchio.
Collapse
Affiliation(s)
- Paula J Bates
- Department of Medicine, University of Louisville, USA; James Graham Brown Cancer Center, University of Louisville, USA.
| | | | - Mohammad T Malik
- Department of Medicine, University of Louisville, USA; James Graham Brown Cancer Center, University of Louisville, USA
| | - Emily M Murphy
- Department of Biomedical Engineering, University of Louisville, USA
| | - Martin G O'Toole
- Department of Biomedical Engineering, University of Louisville, USA
| | - John O Trent
- Department of Medicine, University of Louisville, USA; James Graham Brown Cancer Center, University of Louisville, USA
| |
Collapse
|
28
|
Haddad Zahmatkesh M, Abedi SM, Hosseinimehr SJ. Preparation and biological evaluation of 99mTc-HYNIC-(Ser)3-D4 peptide for targeting and imaging of non-small-cell lung cancer. Future Oncol 2017; 13:893-905. [DOI: 10.2217/fon-2016-0426] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: In this study, radiolabeled D4 peptide conjugate was studied as a radiotracer for imaging of non-small-cell lung cancer with overexpression of EGFR. Methods: HYNIC-(Ser)3-D4 peptide was labeled with 99mTc using tricine as a co-ligand. Cellular specific binding and internalization as well as in vivo tumor targeting were assessed. Results: The in vitro experiments showed good cellular specific binding. Tumor uptake values as %ID/g were 7.55 and 6.82% at 1 and 4 h after injection, respectively. The presaturation of EGFR in xenografted nude mice reduced 36% tumor uptake of radioactivity at 1 h after injection that confirmed in vivo specificity. Conclusion: Findings showed this radiolabeled peptide is a promising candidate for tumor targeting and molecular imaging of non-small-cell lung cancer.
Collapse
Affiliation(s)
- Mona Haddad Zahmatkesh
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mohammad Abedi
- Department of Radiology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Cardiovascular Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
29
|
de Sousa Lacerda CM, Ferreira IM, Dos Santos SR, de Barros ALB, Fernandes SO, Cardoso VN, de Andrade ASR. (1→3)-β-D-glucan aptamers labeled with technetium-99m: Biodistribution and imaging in experimental models of bacterial and fungal infection. Nucl Med Biol 2016; 46:19-24. [PMID: 27951452 DOI: 10.1016/j.nucmedbio.2016.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/18/2016] [Accepted: 11/24/2016] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Acid nucleic aptamers are RNA or DNA oligonucleotides capable of binding to a target molecule with high affinity and selectivity. These molecules are promising tools in nuclear medicine. Many aptamers have been used as targeting molecule of radiopharmaceuticals in preclinical studies. (1→3)-β-D-glucans are the main structural cell wall components of fungi and some bacteria. In the present study two radiolabeled (1→3)-β-D-glucan aptamers (seq6 and seq30) were evaluated to identity infectious foci caused by fungal or bacterial cells. METHODS Aptamer labeling with 99mTc was performed by the direct method and biodistribution studies were accomplished in Swiss mice (n=6) infected in the right thigh muscle with Staphylococcus aureus or Candida albicans. A 99mTc radiolabeled library consisting of oligonucleotides with random sequences was used as control. RESULTS There was a higher uptake of 99mTc radiolabeled aptamers in the infected thigh than in the left thigh muscle (non-infected) in the S. aureus infected animals. The target/non-target ratios were 3.17±0.22 for seq6 and 2.66±0.10 for seq30. These ratios were statistically higher than the value (1.54±0.05) found for the radiolabeled library (control). With regard to biodistribution, no statistical difference was verified between aptamers and control uptakes in the infection foci in the C. albicans infected animals. The target/non-target ratios were 1.53±0.03, 1.64±0.12 and 1.08±0.02 for radiolabeled library, seq6 and seq30, respectively. Scintigraphic imaging of infected foci using radiolabeled aptamers was possible only for S. aureus infected mice. CONCLUSIONS Seq6 and seq30 aptamers proved to be inefficient for diagnosis of C. albicans infection. Nevertheless, their applicability for diagnosis of S. aureus and other bacterial infections by scintigraphy should be further explored.
Collapse
Affiliation(s)
- Camila Maria de Sousa Lacerda
- Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Rua Professor Mário Werneck S/N°, Cidade Universitária-Campus da UFMG, Belo Horizonte, MG 31120-970, Brazil.
| | - Iêda Mendes Ferreira
- Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Rua Professor Mário Werneck S/N°, Cidade Universitária-Campus da UFMG, Belo Horizonte, MG 31120-970, Brazil.
| | - Sara Roberta Dos Santos
- Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Rua Professor Mário Werneck S/N°, Cidade Universitária-Campus da UFMG, Belo Horizonte, MG 31120-970, Brazil.
| | - André Luís Branco de Barros
- Departamento de Análises Clínicas e Toxicológicas-Faculdade de Farmácia, Universidade Federal de Minas Gerais, Cidade Universitária-Campus da UFMG, Belo Horizonte, MG 31270-091, Brazil.
| | - Simone Odília Fernandes
- Departamento de Análises Clínicas e Toxicológicas-Faculdade de Farmácia, Universidade Federal de Minas Gerais, Cidade Universitária-Campus da UFMG, Belo Horizonte, MG 31270-091, Brazil.
| | - Valbert Nascimento Cardoso
- Departamento de Análises Clínicas e Toxicológicas-Faculdade de Farmácia, Universidade Federal de Minas Gerais, Cidade Universitária-Campus da UFMG, Belo Horizonte, MG 31270-091, Brazil.
| | - Antero Silva Ribeiro de Andrade
- Centro de Desenvolvimento da Tecnologia Nuclear (CDTN), Rua Professor Mário Werneck S/N°, Cidade Universitária-Campus da UFMG, Belo Horizonte, MG 31120-970, Brazil.
| |
Collapse
|
30
|
Abedi SM, Mardanshahi A, Shahhosseini R, Hosseinimehr SJ. Nuclear medicine for imaging of epithelial ovarian cancer. Future Oncol 2016; 12:1165-77. [PMID: 26984362 DOI: 10.2217/fon.16.19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cancer is one of the leading causes of mortality worldwide. Usually, the diagnosis of cancer at an early stage is important to facilitate proper treatment and survival. Nuclear medicine has been successfully used in the diagnosis, staging, therapy and monitoring of cancers. Single-photon emission computed tomography and PET-based companion imaging agents are in development for use as a companion diagnostic tool for patients with ovarian cancer. The present review discusses the basic and clinical studies related to the use of radiopharmaceuticals in the diagnosis and management of ovarian cancer, focusing on their utility and comparing them with other imaging techniques such as computed tomography and MRI.
Collapse
Affiliation(s)
- Seyed Mohammad Abedi
- Department of Radiology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Mardanshahi
- Department of Radiology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Roza Shahhosseini
- Department of Obstetrics & Gynecology, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.,Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
31
|
Kryza D, Debordeaux F, Azéma L, Hassan A, Paurelle O, Schulz J, Savona-Baron C, Charignon E, Bonazza P, Taleb J, Fernandez P, Janier M, Toulmé JJ. Ex Vivo and In Vivo Imaging and Biodistribution of Aptamers Targeting the Human Matrix MetalloProtease-9 in Melanomas. PLoS One 2016; 11:e0149387. [PMID: 26901393 PMCID: PMC4762761 DOI: 10.1371/journal.pone.0149387] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 02/01/2016] [Indexed: 02/07/2023] Open
Abstract
The human Matrix MetalloProtease-9 (hMMP-9) is overexpressed in tumors where it promotes the release of cancer cells thus contributing to tumor metastasis. We raised aptamers against hMMP-9, which constitutes a validated marker of malignant tumors, in order to design probes for imaging tumors in human beings. A chemically modified RNA aptamer (F3B), fully resistant to nucleases was previously described. This compound was subsequently used for the preparation of F3B-Cy5, F3B-S-acetylmercaptoacetyltriglycine (MAG) and F3B-DOTA. The binding properties of these derivatives were determined by surface plasmon resonance and electrophoretic mobility shift assay. Optical fluorescence imaging confirmed the binding to hMMP-9 in A375 melanoma bearing mice. Quantitative biodistribution studies were performed at 30 min, 1h and 2 h post injection of 99mTc-MAG-aptamer and 111In-DOTA-F3B. 99mTc radiolabeled aptamer specifically detected hMMP-9 in A375 melanoma tumors but accumulation in digestive tract was very high. Following i.v. injection of 111In-DOTA-F3B, high level of radioactivity was observed in kidneys and bladder but digestive tract uptake was very limited. Tumor uptake was significantly (student t test, p<0.05) higher for 111In-DOTA-F3B with 2.0%ID/g than for the 111In-DOTA-control oligonucleotide (0.7%ID/g) with tumor to muscle ratio of 4.0. Such difference in tumor accumulation has been confirmed by ex vivo scintigraphic images performed at 1h post injection and by autoradiography, which revealed the overexpression of hMMP-9 in sections of human melanomas. These results demonstrate that F3B aptamer is of interest for detecting hMMP-9 in melanoma tumor.
Collapse
Affiliation(s)
- David Kryza
- UNIV Lyon, Université Claude Bernard Lyon 1, LAGEP UMR 5007 CNRS, Villeurbanne, France
- Hospices Civils de Lyon, Imthernat plateform, Lyon, France
| | - Frédéric Debordeaux
- Université de Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France
- Service de Médecine Nucléaire, CHU de Bordeaux, Bordeaux, France
| | - Laurent Azéma
- Inserm U1212, Bordeaux, France
- CNRS UMR5320, Pessac, France
- University of Bordeaux, ARNA Laboratory, Bordeaux, France
| | - Aref Hassan
- Inserm U1212, Bordeaux, France
- CNRS UMR5320, Pessac, France
- University of Bordeaux, ARNA Laboratory, Bordeaux, France
| | - Olivier Paurelle
- Inserm U1212, Bordeaux, France
- CNRS UMR5320, Pessac, France
- University of Bordeaux, ARNA Laboratory, Bordeaux, France
| | - Jürgen Schulz
- Université de Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France
- Service de Médecine Nucléaire, CHU de Bordeaux, Bordeaux, France
| | | | - Elsa Charignon
- Hospices Civils de Lyon, Imthernat plateform, Lyon, France
| | | | - Jacqueline Taleb
- UNIV Lyon, Université Claude Bernard Lyon 1, LAGEP UMR 5007 CNRS, Villeurbanne, France
| | - Philippe Fernandez
- Université de Bordeaux, CNRS, INCIA, UMR 5287, Bordeaux, France
- Service de Médecine Nucléaire, CHU de Bordeaux, Bordeaux, France
| | - Marc Janier
- UNIV Lyon, Université Claude Bernard Lyon 1, LAGEP UMR 5007 CNRS, Villeurbanne, France
- Hospices Civils de Lyon, Imthernat plateform, Lyon, France
| | - Jean Jacques Toulmé
- Inserm U1212, Bordeaux, France
- CNRS UMR5320, Pessac, France
- University of Bordeaux, ARNA Laboratory, Bordeaux, France
| |
Collapse
|