1
|
Yang C, Jiang Y, Hao SH, Yan XY, Hong DF, Naranmandura H. Aptamers: an emerging navigation tool of therapeutic agents for targeted cancer therapy. J Mater Chem B 2021; 10:20-33. [PMID: 34881767 DOI: 10.1039/d1tb02098f] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chemotherapeutic agents have been used for the treatment of numerous cancers, but due to poor selectivity and severe systemic side effects, their clinical application is limited. Single-stranded DNA (ssDNA) or RNA aptamers could conjugate with highly toxic chemotherapy drugs, toxins, therapeutic RNAs or other molecules as novel aptamer-drug conjugates (ApDCs), which are capable of significantly improving the therapeutic efficacy and reducing the systemic toxicity of drugs and have great potential in clinics for targeted cancer therapy. In this review, we have comprehensively discussed and summarized the current advances in the screening approaches of aptamers for specific cancer biomarker targeting and development of the aptamer-drug conjugate strategy for targeted drug delivery. Moreover, considering the huge progress in artificial intelligence (AI) for protein and RNA structure predictions, automatic design of aptamers using deep/machine learning techniques could be a powerful approach for rapid and precise construction of biopharmaceutics (i.e., ApDCs) for application in cancer targeted therapy.
Collapse
Affiliation(s)
- Chang Yang
- Department of Hematology, the First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China. .,Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, China
| | - Yu Jiang
- Department of Hematology, the First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China. .,Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sai Heng Hao
- College of Pharmaceutical Sciences, Inner Mongolia Medical University, Hohhot, China
| | - Xing Yi Yan
- Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, China.,Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China
| | - De Fei Hong
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Hua Naranmandura
- Department of Hematology, the First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, China. .,Department of Toxicology, School of Medicine and Public Health, Zhejiang University, Hangzhou, China.,Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang University Cancer Center, Hangzhou, China
| |
Collapse
|
2
|
Ge MH, Zhu XH, Shao YM, Wang C, Huang P, Wang Y, Jiang Y, Maimaitiyiming Y, Chen E, Yang C, Naranmandura H. Synthesis and characterization of CD133 targeted aptamer-drug conjugates for precision therapy of anaplastic thyroid cancer. Biomater Sci 2021; 9:1313-1324. [PMID: 33350399 DOI: 10.1039/d0bm01832e] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Anaplastic thyroid cancer (ATC) is an undifferentiated and highly aggressive type of thyroid cancer and is extremely resistant to standard therapies such as surgical resection and radioactive iodine therapy. Although targeted therapeutic agents including small molecule drugs and monoclonal antibodies are rapidly developed in recent years, no ATC targeted drugs are available to date; thereby, novel targeted therapies are needed to improve the outcomes of ATC patients. Aptamers are single-stranded DNA (or RNA) molecules that can selectively bind to cancer specific antigens, and aptamer-based targeted therapy has certain advantages over that based on antibodies due to its high binding affinity and low immunogenicity. Here, we identified that CD133, a cancer stem cell marker, was specifically expressed in ATC tumor tissues and cells, implying that CD133 is a potential drug target for ATC therapy. Additionally, we successfully obtained a CD133 targeted aptamer AP-1 by paired cell-based SELEX, which can precisely recognize CD133 antigen in vitro. Furthermore, the truncated AP-1-M aptamer from its precursor AP-1 has shown higher binding affinity for CD133, and specifically accumulated in anaplastic thyroid cancer FRO cell derived tumor in vivo. Conjugation of truncated AP-1-M with doxorubicin could dramatically inhibit CD133 positive FRO cell proliferation, induce cell apoptosis in vitro, and also suppress tumor growth in FRO cell xenograft mice in vivo. Our results clearly demonstrated that the CD133 targeted aptamer AP-1-M conjugated with anticancer drugs has potential to become a promising therapeutic approach against ATC in the near future.
Collapse
Affiliation(s)
- Ming Hua Ge
- Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Xu Hang Zhu
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yi Ming Shao
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Chao Wang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Ping Huang
- Department of Pharmacy, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Yun Wang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Yu Jiang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou 310058, China.
| | - Yasen Maimaitiyiming
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou 310058, China. and Department of Public Health, and Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - En Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chang Yang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou 310058, China. and Department of Public Health, and Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hua Naranmandura
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou 310058, China. and Department of Public Health, and Department of Hematology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China and Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
| |
Collapse
|
3
|
Pang B, Yang H, Wang L, Chen J, Jin L, Shen B. Aptamer modified MoS2 nanosheets application in targeted photothermal therapy for breast cancer. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2020.125506] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
4
|
Yazdian-Robati R, Bayat P, Oroojalian F, Zargari M, Ramezani M, Taghdisi SM, Abnous K. Therapeutic applications of AS1411 aptamer, an update review. Int J Biol Macromol 2020; 155:1420-1431. [PMID: 31734366 DOI: 10.1016/j.ijbiomac.2019.11.118] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023]
Abstract
Nucleolin or C23, is one of the most abundant non-ribosomal phosphoproteins of nucleolus. However, in several cancers, nucleolin is highly expressed both intracellularly and on the cell surface. So, it is considered as a potential target for the diagnosis and cancer therapy. Targeting nucleolin by compounds such as AS1411 aptamer can reduce tumor cell growth. In this regard, interest has increased in nucleolin as a molecular target for overcoming cancer therapy challenges. This review paper addressed recent progresses in nucleolin targeting by the G-rich AS1411 aptamer in the field of cancer therapy mainly over the past three years.
Collapse
Affiliation(s)
- Rezvan Yazdian-Robati
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Payam Bayat
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehryar Zargari
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
5
|
Development of molecularly imprinted magnetic iron oxide nanoparticles for doxorubicin drug delivery. MONATSHEFTE FUR CHEMIE 2020. [DOI: 10.1007/s00706-020-02644-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
6
|
Zhang L, Wang S, Yang Z, Hoshika S, Xie S, Li J, Chen X, Wan S, Li L, Benner SA, Tan W. An Aptamer-Nanotrain Assembled from Six-Letter DNA Delivers Doxorubicin Selectively to Liver Cancer Cells. Angew Chem Int Ed Engl 2019; 59:663-668. [PMID: 31650689 DOI: 10.1002/anie.201909691] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/16/2019] [Indexed: 12/30/2022]
Abstract
Expanding the number of nucleotides in DNA increases the information density of functional DNA molecules, creating nanoassemblies that cannot be invaded by natural DNA/RNA in complex biological systems. Here, we show how six-letter GACTZP DNA contributes this property in two parts of a nanoassembly: 1) in an aptamer evolved from a six-letter DNA library to selectively bind liver cancer cells; and 2) in a six-letter self-assembling GACTZP nanotrain that carries the drug doxorubicin. The aptamer-nanotrain assembly, charged with doxorubicin, selectively kills liver cancer cells in culture, as the selectivity of the aptamer binding directs doxorubicin into the aptamer-targeted cells. The assembly does not kill untransformed cells that the aptamer does not bind. This architecture, built with an expanded genetic alphabet, is reminiscent of antibodies conjugated to drugs, which presumably act by this mechanism as well, but with the antibody replaced by an aptamer.
Collapse
Affiliation(s)
- Liqin Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China.,Department of Chemistry, Department of Physiology and Functional Genomics, UF Health Cancer Center, UF Genetics Institute, University of Florida, Gainesville, FL, 32611, USA
| | - Sai Wang
- Department of Chemistry, Department of Physiology and Functional Genomics, UF Health Cancer Center, UF Genetics Institute, University of Florida, Gainesville, FL, 32611, USA.,Current address: College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, 266003, China
| | - Zunyi Yang
- Foundation for Applied Molecular Evolution, 13709 Progress Boulevard, Box 7, Alachua, FL, 32615, USA.,Firebird Biomolecular Sciences LLC, 13709 Progress Boulevard, Box 17, Alachua, FL, 32615, USA
| | - Shuichi Hoshika
- Foundation for Applied Molecular Evolution, 13709 Progress Boulevard, Box 7, Alachua, FL, 32615, USA.,Firebird Biomolecular Sciences LLC, 13709 Progress Boulevard, Box 17, Alachua, FL, 32615, USA
| | - Sitao Xie
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Jin Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Xigao Chen
- Department of Chemistry, Department of Physiology and Functional Genomics, UF Health Cancer Center, UF Genetics Institute, University of Florida, Gainesville, FL, 32611, USA
| | - Shuo Wan
- Department of Chemistry, Department of Physiology and Functional Genomics, UF Health Cancer Center, UF Genetics Institute, University of Florida, Gainesville, FL, 32611, USA
| | - Long Li
- Department of Chemistry, Department of Physiology and Functional Genomics, UF Health Cancer Center, UF Genetics Institute, University of Florida, Gainesville, FL, 32611, USA
| | - Steven A Benner
- Foundation for Applied Molecular Evolution, 13709 Progress Boulevard, Box 7, Alachua, FL, 32615, USA.,Firebird Biomolecular Sciences LLC, 13709 Progress Boulevard, Box 17, Alachua, FL, 32615, USA
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China.,Department of Chemistry, Department of Physiology and Functional Genomics, UF Health Cancer Center, UF Genetics Institute, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
7
|
Zhang L, Wang S, Yang Z, Hoshika S, Xie S, Li J, Chen X, Wan S, Li L, Benner SA, Tan W. An Aptamer‐Nanotrain Assembled from Six‐Letter DNA Delivers Doxorubicin Selectively to Liver Cancer Cells. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201909691] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Liqin Zhang
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering College of Biology, Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
- Department of Chemistry Department of Physiology and Functional Genomics UF Health Cancer Center UF Genetics Institute University of Florida Gainesville FL 32611 USA
| | - Sai Wang
- Department of Chemistry Department of Physiology and Functional Genomics UF Health Cancer Center UF Genetics Institute University of Florida Gainesville FL 32611 USA
- Current address: College of Food Science and Engineering Ocean University of China Qingdao Shandong 266003 China
| | - Zunyi Yang
- Foundation for Applied Molecular Evolution 13709 Progress Boulevard, Box 7 Alachua FL 32615 USA
- Firebird Biomolecular Sciences LLC 13709 Progress Boulevard, Box 17 Alachua FL 32615 USA
| | - Shuichi Hoshika
- Foundation for Applied Molecular Evolution 13709 Progress Boulevard, Box 7 Alachua FL 32615 USA
- Firebird Biomolecular Sciences LLC 13709 Progress Boulevard, Box 17 Alachua FL 32615 USA
| | - Sitao Xie
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering College of Biology, Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Jin Li
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering College of Biology, Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
| | - Xigao Chen
- Department of Chemistry Department of Physiology and Functional Genomics UF Health Cancer Center UF Genetics Institute University of Florida Gainesville FL 32611 USA
| | - Shuo Wan
- Department of Chemistry Department of Physiology and Functional Genomics UF Health Cancer Center UF Genetics Institute University of Florida Gainesville FL 32611 USA
| | - Long Li
- Department of Chemistry Department of Physiology and Functional Genomics UF Health Cancer Center UF Genetics Institute University of Florida Gainesville FL 32611 USA
| | - Steven A. Benner
- Foundation for Applied Molecular Evolution 13709 Progress Boulevard, Box 7 Alachua FL 32615 USA
- Firebird Biomolecular Sciences LLC 13709 Progress Boulevard, Box 17 Alachua FL 32615 USA
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL) State Key Laboratory of Chemo/Biosensing and Chemometrics College of Chemistry and Chemical Engineering College of Biology, Aptamer Engineering Center of Hunan Province Hunan University Changsha Hunan 410082 China
- Department of Chemistry Department of Physiology and Functional Genomics UF Health Cancer Center UF Genetics Institute University of Florida Gainesville FL 32611 USA
| |
Collapse
|
8
|
Roki N, Tsinas Z, Solomon M, Bowers J, Getts RC, Muro S. Unprecedently high targeting specificity toward lung ICAM-1 using 3DNA nanocarriers. J Control Release 2019; 305:41-49. [PMID: 31100312 PMCID: PMC7171557 DOI: 10.1016/j.jconrel.2019.05.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/05/2019] [Accepted: 05/13/2019] [Indexed: 12/17/2022]
Abstract
DNA nanostructures hold great potential for drug delivery. However, their specific targeting is often compromised by recognition by scavenger receptors involved in clearance. In our previous study in cell culture, we showed targeting specificity of a 180 nm, 4-layer DNA-built nanocarrier called 3DNA coupled with antibodies against intercellular adhesion molecule-1 (ICAM-1), a glycoprotein overexpressed in the lungs in many diseases. Here, we examined the biodistribution of various 3DNA formulations in mice. A formulation consisted of 3DNA whose outer-layer arms were hybridized to secondary antibody-oligonucleotide conjugates. Anchoring IgG on this formulation reduced circulation and kidney accumulation vs. non-anchored IgG, while increasing liver and spleen clearance, as expected for a nanocarrier. Anchoring anti-ICAM changed the biodistribution of this antibody similarly, yet this formulation specifically accumulated in the lungs, the main ICAM-1 target. Since lung targeting was modest (2-fold specificity index over IgG formulation), we pursued a second preparation involving direct hybridization of primary antibody-oligonucleotide conjugates to 3DNA. This formulation had prolonged stability in serum and showed a dramatic increase in lung distribution: the specificity index was 424-fold above a matching IgG formulation, 144-fold more specific than observed for PLGA nanoparticles of similar size, polydispersity, ζ-potential and antibody valency, and its lung accumulation increased with the number of anti-ICAM molecules per particle. Immunohistochemistry showed that anti-ICAM and 3DNA components colocalized in the lungs, specifically associating with endothelial markers, without apparent histological changes. The degree of in vivo targeting for anti-ICAM/3DNA-nanocarriers is unprecedented, for which this platform technology holds great potential to develop future therapeutic applications.
Collapse
Affiliation(s)
- Nikša Roki
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Zois Tsinas
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Melani Solomon
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA
| | | | | | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, MD, USA; Institute for Bioengineering of Catalonia of the Barcelona Institute of Science and Technology, Institution of Catalonia for Research and Advanced Studies, Barcelona, Spain.
| |
Collapse
|
9
|
Yang C, Wang Y, Ge MH, Fu YJ, Hao R, Islam K, Huang P, Chen F, Sun J, Hong DF, Naranmandura H. Rapid identification of specific DNA aptamers precisely targeting CD33 positive leukemia cells through a paired cell-based approach. Biomater Sci 2019; 7:938-950. [PMID: 30519686 DOI: 10.1039/c8bm01393d] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Aptamers are short single-stranded DNA or RNA molecules, which have recently been developed for potential broad applications such as clinical therapeutics, diagnosis and tumor-targeted drug delivery. However, the selection of specific aptamers is often unsatisfactory using the classical protein or cell-based SELEX. Herein, we modified the paired cell line approach to identify aptamers targeting leukemia cells expressing the CD33 antigen. Our strategy artfully used the same cells for negative (HEK293T cells) and positive (CD33 transfected-HEK293T cells) aptamer selections, and the negative selections were performed adequately before the positive selection to remove unspecific sequences. The advantages of this strategy are that it is fast and accurate, where only a few rounds of selection together with PCR amplifications are sufficient to obtain high binding affinity antigen-targeted aptamers. By using our modified approach, we successfully obtained the CD33-targeting aptamer S30, which could highly recognize the C2 domain of the CD33 antigen in vitro and in vivo. Moreover, the optimized aptamer S30-T1 (i.e., core region of S30) was conjugated with doxorubicin (Dox) to synthesize S30-T1-Dox conjugates, which could specifically inhibit CD33 positive acute myeloid leukemia HL-60 cell proliferation by arresting the cell cycle at the G2 phase. Thus, our modified approach can rapidly screen reliable, stable and high binding affinity aptamers for precise cancer treatment.
Collapse
Affiliation(s)
- Chang Yang
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou 310058, P.R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Liu Q, Wang D, Xu Z, Huang C, Zhang C, He B, Mao C, Wang G, Qian H. Targeted Delivery of Rab26 siRNA with Precisely Tailored DNA Prism for Lung Cancer Therapy. Chembiochem 2019; 20:1139-1144. [PMID: 30610755 DOI: 10.1002/cbic.201800761] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Indexed: 01/06/2023]
Abstract
Programmable DNA nanostructures are a new class of biocompatible, nontoxic nanomaterials. Nevertheless, their application in the field of biomedical research is still in its infancy, especially as drug delivery vehicles for gene therapy. In this study, a GTPase Rab26 was investigated as a new potential therapeutic target using a precisely tailored DNA nanoprism for targeted lung cancer therapy. Specifically, a DNA nanoprism platform with tunable targeting and siRNA loading capability is designed and synthesized. The as-prepared DNA prisms were decorated with two functional units: a Rab26 siRNA as the drug and MUC-1 aptamers as a targeting moiety for non-small cell lung cancer. The number and position of both siRNA and MUC-1 aptamers can be readily tuned by switching two short, single-stranded DNA. Native polyacrylamide gel electrophoresis (PAGE) and dynamic light scattering technique (DLS) demonstrate that all nanoprisms with different functionalities are self-assembled with high yield. It is also found that the cellular uptake of DNA prisms is proportional to the aptamer number on each nanoprism, and the as-prepared DNA nanoprism show excellent anti-cancer activities and targeting capability. This study suggests that by careful design, self-assembled DNA nanostructures are highly promising, customizable, multifunctional nanoplatforms for potential biomedical applications, such as personalized precision therapy.
Collapse
Affiliation(s)
- Qian Liu
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Street, Chongqing, 400037, China
| | - Dong Wang
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Street, Chongqing, 400037, China
| | - Zhi Xu
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Street, Chongqing, 400037, China
| | - Chunji Huang
- Basic Medical College, Third Military Medical University, Chongqing, 400038, China
| | - Chun Zhang
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Street, Chongqing, 400037, China
| | - Binfeng He
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Street, Chongqing, 400037, China
| | - Chengde Mao
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN, 47907, USA
| | - Guansong Wang
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Street, Chongqing, 400037, China
| | - Hang Qian
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Street, Chongqing, 400037, China
| |
Collapse
|
11
|
Yazdian-Robati R, Arab A, Ramezani M, Rafatpanah H, Bahreyni A, Nabavinia MS, Abnous K, Taghdisi SM. Smart aptamer-modified calcium carbonate nanoparticles for controlled release and targeted delivery of epirubicin and melittin into cancer cells in vitro and in vivo. Drug Dev Ind Pharm 2019; 45:603-610. [PMID: 30633594 DOI: 10.1080/03639045.2019.1569029] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
To explore the effect of combination therapy of epirubicin (Epi) and melittin (Mel) to cancer cells, calcium carbonate nanoparticles (CCN), as carriers, were developed which were modified with MUC1-Dimer aptamers as targeting agents. Both Epi and Mel were delivered at the same time to cancer cells overexpressing the target of MUC1 aptamer, mucin 1 glycoproteins (MCF7 and C26 cells). CCN were prepared with a water-in-oil emulsion method. Epi and Mel were separately encapsulated in CCN and the nanoparticles were modified with MUC1-Dimer aptamers. In vitro studies, including MTT assay, flow cytometry analysis and fluorescence imaging were applied to investigate the targeting and cell proliferation inhibition capabilities of MUC1-Dimer aptamer-CCN-Mel complex and MUC1-Dimer aptamer-CCN-Epi complex in the target (MCF-7 and C26 cells) and nontarget (HepG2) cells. Also, the function of the developed complexes was analyzed using in vivo tumor growth inhibition. The release of Epi from MUC1-Dimer aptamer-CCN-Epi complex was pH-sensitive. Cellular uptake studies showed more internalization of the MUC1-Dimer aptamer-CCN-Epi complex into MCF-7 and C26 cells (target) compared to HepG2 cells (nontarget). Interestingly, the MUC1-Dimer aptamer-CCN-Mel complex and MUC1-Dimer aptamer-CCN-Epi complex indicated very low toxicity as compared to target cells. Moreover, co-delivery of Epi and Mel using the mixture of MUC1-Dimer aptamer-CCN-Mel complex and MUC1-Dimer aptamer-CCN-Epi complex exhibited strong synergistic cytotoxicity in MCF-7 and C26 cells. Furthermore, the presented complexes had a better function to control tumor growth in vivo compared to free Epi.
Collapse
Affiliation(s)
- Rezvan Yazdian-Robati
- a Molecular and Cell biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences , Sari , Iran
| | - Atefeh Arab
- b Department of Pharmaceutical Biotechnology , School of Pharmacy, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Mohammad Ramezani
- c Pharmaceutical Research Center , Pharmaceutical Technology Institute, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Houshang Rafatpanah
- d Faculty of Medicine, Department of Immunology , Immunology Research Center, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Amirhossein Bahreyni
- c Pharmaceutical Research Center , Pharmaceutical Technology Institute, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Maryam Sadat Nabavinia
- e Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Department of Pharmacognosy , Shahid Sadoughi University of Medical Sciences , Yazd , Iran
| | - Khalil Abnous
- c Pharmaceutical Research Center , Pharmaceutical Technology Institute, Mashhad University of Medical Sciences , Mashhad , Iran
| | - Seyed Mohammad Taghdisi
- f Targeted Drug Delivery Research Center , Pharmaceutical Technology Institute, Mashhad University of Medical Sciences , Mashhad , Iran
| |
Collapse
|
12
|
Pusuluri A, Krishnan V, Lensch V, Sarode A, Bunyan E, Vogus DR, Menegatti S, Soh HT, Mitragotri S. Treating Tumors at Low Drug Doses Using an Aptamer-Peptide Synergistic Drug Conjugate. Angew Chem Int Ed Engl 2018; 58:1437-1441. [PMID: 30537284 DOI: 10.1002/anie.201812650] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Indexed: 11/06/2022]
Abstract
Combination chemotherapy must strike a difficult balance between safety and efficacy. Current regimens suffer from poor therapeutic impact because drugs are given at their maximum tolerated dose (MTD), which compounds the toxicity risk and exposes tumors to non-optimal drug ratios. A modular framework has been developed that selectively delivers drug combinations at synergistic ratios via tumor-targeting aptamers for effective low-dose treatment. A nucleolin-recognizing aptamer was coupled to peptide scaffolds laden with precise ratios of doxorubicin (DOX) and camptothecin (CPT). This construct had an extremely low IC50 (31.9 nm) against MDA-MB-231 breast cancer cells in vitro, and exhibited in vivo efficacy at micro-dose injections (500 and 350 μg kg-1 dose-1 of DOX and CPT, respectively) that are 20-30-fold lower than their previously-reported MTDs. This approach represents a generalizable strategy for the safe and consistent delivery of combination drugs in oncology.
Collapse
Affiliation(s)
- Anusha Pusuluri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.,Wyss Institute of Biologically Inspired Engineering, Harvard University, Harvard University, Boston, MA, 02115, USA.,Department of Chemical Engineering, University of California, Santa Barbara, CA, 93106, USA
| | - Vinu Krishnan
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.,Wyss Institute of Biologically Inspired Engineering, Harvard University, Harvard University, Boston, MA, 02115, USA
| | - Valerie Lensch
- Department of Chemical Engineering, University of California, Santa Barbara, CA, 93106, USA
| | - Apoorva Sarode
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.,Wyss Institute of Biologically Inspired Engineering, Harvard University, Harvard University, Boston, MA, 02115, USA
| | - Elaine Bunyan
- Department of Chemical Engineering, University of California, Santa Barbara, CA, 93106, USA
| | - Douglas R Vogus
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.,Wyss Institute of Biologically Inspired Engineering, Harvard University, Harvard University, Boston, MA, 02115, USA
| | - Stefano Menegatti
- Department of Chemical & Biomolecular Engineering, North Carolina State University, Raleigh, NC, 27606, USA
| | - H Tom Soh
- Department of Electrical Engineering and Department of Radiology, Stanford University, Palo Alto, CA, 94305, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.,Wyss Institute of Biologically Inspired Engineering, Harvard University, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
13
|
Pusuluri A, Krishnan V, Lensch V, Sarode A, Bunyan E, Vogus DR, Menegatti S, Soh HT, Mitragotri S. Treating Tumors at Low Drug Doses Using an Aptamer–Peptide Synergistic Drug Conjugate. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201812650] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Anusha Pusuluri
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering, Harvard University Harvard University Boston MA 02115 USA
- Department of Chemical Engineering University of California Santa Barbara CA 93106 USA
| | - Vinu Krishnan
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering, Harvard University Harvard University Boston MA 02115 USA
| | - Valerie Lensch
- Department of Chemical Engineering University of California Santa Barbara CA 93106 USA
| | - Apoorva Sarode
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering, Harvard University Harvard University Boston MA 02115 USA
| | - Elaine Bunyan
- Department of Chemical Engineering University of California Santa Barbara CA 93106 USA
| | - Douglas R. Vogus
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering, Harvard University Harvard University Boston MA 02115 USA
| | - Stefano Menegatti
- Department of Chemical & Biomolecular Engineering North Carolina State University Raleigh NC 27606 USA
| | - H. Tom Soh
- Department of Electrical Engineering and Department of Radiology Stanford University Palo Alto CA 94305 USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences Harvard University Cambridge MA 02138 USA
- Wyss Institute of Biologically Inspired Engineering, Harvard University Harvard University Boston MA 02115 USA
| |
Collapse
|
14
|
Shi L, Mu C, Gao T, Chen T, Hei S, Yang J, Li G. DNA nanoflower blooms in nanochannels: a new strategy for miRNA detection. Chem Commun (Camb) 2018; 54:11391-11394. [DOI: 10.1039/c8cc05690k] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
DNA nanoflower blooming was employed in the nanochannels of porous anodic alumina to build a nanochannel platform for miRNA detection with excellent sensitivity, selectivity and reproducibility.
Collapse
Affiliation(s)
- Liu Shi
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences
- Department of Biochemistry
- Nanjing University
- Nanjing 210093
- P. R. China
| | - Chaoli Mu
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences
- Department of Biochemistry
- Nanjing University
- Nanjing 210093
- P. R. China
| | - Tao Gao
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences
- Department of Biochemistry
- Nanjing University
- Nanjing 210093
- P. R. China
| | - Tianshu Chen
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- P. R. China
| | - Shuang Hei
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences
- Department of Biochemistry
- Nanjing University
- Nanjing 210093
- P. R. China
| | - Jie Yang
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences
- Department of Biochemistry
- Nanjing University
- Nanjing 210093
- P. R. China
| | - Genxi Li
- State Key Laboratory of Pharmaceutical Biotechnology and Collaborative Innovation Center of Chemistry for Life Sciences
- Department of Biochemistry
- Nanjing University
- Nanjing 210093
- P. R. China
| |
Collapse
|
15
|
Yang F, Zhang TT, Li SS, Song P, Zhang K, Guan QY, Kang B, Xu JJ, Chen HY. Endogenous MicroRNA-Triggered and Real-Time Monitored Drug Release via Cascaded Energy Transfer Payloads. Anal Chem 2017; 89:10239-10247. [DOI: 10.1021/acs.analchem.7b01582] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Fan Yang
- State Key Laboratory of Analytical
Chemistry for Life Science and Collaborative Innovation Center of
Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Ting-Ting Zhang
- State Key Laboratory of Analytical
Chemistry for Life Science and Collaborative Innovation Center of
Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Shan-Shan Li
- State Key Laboratory of Analytical
Chemistry for Life Science and Collaborative Innovation Center of
Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Pei Song
- State Key Laboratory of Analytical
Chemistry for Life Science and Collaborative Innovation Center of
Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Kai Zhang
- State Key Laboratory of Analytical
Chemistry for Life Science and Collaborative Innovation Center of
Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Qi-Yuan Guan
- State Key Laboratory of Analytical
Chemistry for Life Science and Collaborative Innovation Center of
Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Bin Kang
- State Key Laboratory of Analytical
Chemistry for Life Science and Collaborative Innovation Center of
Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jing-Juan Xu
- State Key Laboratory of Analytical
Chemistry for Life Science and Collaborative Innovation Center of
Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Hong-Yuan Chen
- State Key Laboratory of Analytical
Chemistry for Life Science and Collaborative Innovation Center of
Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
16
|
Ding Q, Zhan Q, Zhou X, Zhang T, Xing D. Theranostic Upconversion Nanobeacons for Tumor mRNA Ratiometric Fluorescence Detection and Imaging-Monitored Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:5944-5953. [PMID: 27647762 DOI: 10.1002/smll.201601724] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/18/2016] [Indexed: 06/06/2023]
Abstract
Remote optical detection and imaging of specific tumor-related biomarkers and simultaneous activation of therapy according to the expression level of the biomarkers in tumor site with theranostic probes should be an effective modality for treatment of cancers. Herein, an upconversion nanobeacon (UCNPs-MB/Dox) is proposed as a new theranostic nanoprobe to ratiometrically detect and visualize the thymidine kinase 1 (TK1) mRNA that can simultaneously trigger the Dox release to activate the chemotherapy accordingly. UCNPs-MB/Dox is constructed with the conjugation of a TK1 mRNA-specific molecular beacon (MB) bearing a quencher (BHQ-1) and an alkene handle modified upconversion nanoparticle (UCNP) through click reaction and subsequently loading with a chemotherapy drug (Dox). With this nanobeacon, quantitative ratiometric upconversion detection of the target with high sensitivity and selectivity as well as the target triggered Dox release in vitro is demonstrated. The sensitive and selective ratiometric detection and imaging of TK1 mRNA under the irradiation of near infrared light (980 nm) and the mRNA-dependent release of Dox for chemotherapy in the tumor MCF-7 cells and A549 cells are also shown. This work provides a smart and robust platform for gene-related tumor theranostics.
Collapse
Affiliation(s)
- Qianwen Ding
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, P. R. China
| | - Qiuqiang Zhan
- Centre for Optical and Electromagnetic Research, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou, 510006, P. R. China
| | - Xiaoming Zhou
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, P. R. China
| | - Tao Zhang
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, P. R. China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, P. R. China
| |
Collapse
|
17
|
Wang Y, Jiang LP, Zhou S, Bi S, Zhu JJ. DNA Polymerase-Directed Hairpin Assembly for Targeted Drug Delivery and Amplified Biosensing. ACS APPLIED MATERIALS & INTERFACES 2016; 8:26532-26540. [PMID: 27690212 DOI: 10.1021/acsami.6b08597] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Due to the predictable conformation and programmable Watson-Crick base-pairing interactions, DNA has proven to be an attractive material to construct various nanostructures. Herein, we demonstrate a simple model of DNA polymerase-directed hairpin assembly (PDHA) to construct DNA nanoassemblies for versatile applications in biomedicine and biosensing. The system consists of only two hairpins, an initiator and a DNA polymerase. Upon addition of aptamer-linked initiator, the inert stems of the two hairpins are activated alternately under the direction of DNA polymerase, which thus grows into aptamer-tethered DNA nanoassemblies (AptNAs). Moreover, through incorporating fluorophores and drug-loading sites into the AptNAs, we have constructed multifunctional DNA nanoassemblies for targeted cancer therapy with high drug payloads and good biocompatibility. Interestingly, using the as-prepared AptNAs as building blocks, DNA nanohydrogels are self-assembled after centrifugation driven by liquid crystallization and dense packaging of DNA duplexes. Taking advantage of easy preparation and high loading capacity, the PDHAs are readily extended to the fabrication of a label-free biosensing platform, achieving amplified electrochemical detection of microRNA-21 (miR-21) with a detection limit as low as 0.75 fM and a dynamic range of 8 orders of magnitude. This biosensor also demonstrates excellent specificity to discriminate the target miR-21 from the control microRNAs and even the one-base mismatched one and further performs well in analyzing miR-21 in MCF-7 tumor cells.
Collapse
Affiliation(s)
- Yingying Wang
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University , Nanjing 210093, China
| | - Li-Ping Jiang
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University , Nanjing 210093, China
| | - Shiwei Zhou
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University , Nanjing 210093, China
| | - Sai Bi
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University , Nanjing 210093, China
- Collaborative Innovation Center for Marine Biomass Fiber Materials and Textiles, College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Laboratory of Fiber Materials and Modern Textiles, the Growing Base for State Key Laboratory, Qingdao University , Qingdao 266071, China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science and Collaborative Innovation Center of Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University , Nanjing 210093, China
| |
Collapse
|
18
|
Niu W, Chen X, Tan W, Veige AS. N-Heterocyclic Carbene-Gold(I) Complexes Conjugated to a Leukemia-Specific DNA Aptamer for Targeted Drug Delivery. Angew Chem Int Ed Engl 2016; 55:8889-93. [PMID: 27311814 PMCID: PMC5260866 DOI: 10.1002/anie.201602702] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Indexed: 12/23/2022]
Abstract
This report describes the synthesis and characterization of novel N-heterocyclic carbene (NHC)-gold(I) complexes and their bioconjugation to the CCRF-CEM-leukemia-specific aptamer sgc8c. Successful bioconjugation was confirmed by the use of fluorescent tags on both the NHC-Au(I) complex and the aptamer. Cell-viability assays indicated that the NHC-Au(I) -aptamer conjugate was more cytotoxic than the NHC-gold complex alone. A combination of flow cytometry, confocal microscopy, and cell-viability assays provided clear evidence that the NHC-Au(I) -aptamer conjugate was selective for targeted CCRF-CEM leukemia cells.
Collapse
Affiliation(s)
- Weijia Niu
- University of Florida, Department of Chemistry, P.O. Box 117200, Gainesville, FL, USA
| | - Xigao Chen
- University of Florida, Department of Chemistry, P.O. Box 117200, Gainesville, FL, USA
| | - Weihong Tan
- University of Florida, Department of Chemistry, P.O. Box 117200, Gainesville, FL, USA
| | - Adam S Veige
- University of Florida, Department of Chemistry, P.O. Box 117200, Gainesville, FL, USA.
| |
Collapse
|
19
|
Niu W, Chen X, Tan W, Veige AS. N-Heterocyclic Carbene-Gold(I) Complexes Conjugated to a Leukemia-Specific DNA Aptamer for Targeted Drug Delivery. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201602702] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Weijia Niu
- University of Florida; Department of Chemistry; P.O. Box 117200 Gainesville FL USA
| | - Xigao Chen
- University of Florida; Department of Chemistry; P.O. Box 117200 Gainesville FL USA
| | - Weihong Tan
- University of Florida; Department of Chemistry; P.O. Box 117200 Gainesville FL USA
| | - Adam S. Veige
- University of Florida; Department of Chemistry; P.O. Box 117200 Gainesville FL USA
| |
Collapse
|
20
|
Paul M, Dastidar P. Coordination Polymers Derived from Non-Steroidal Anti-Inflammatory Drugs for Cell Imaging and Drug Delivery. Chemistry 2015; 22:988-98. [DOI: 10.1002/chem.201503706] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Indexed: 11/06/2022]
Affiliation(s)
- Mithun Paul
- Department of Organic Chemistry; Indian Association for the Cultivation of Science (IACS); 2A and 2B, Raja S. C. Mullick Road Jadavpur, Kolkata 700032 West Bengal India), Fax
| | - Parthasarathi Dastidar
- Department of Organic Chemistry; Indian Association for the Cultivation of Science (IACS); 2A and 2B, Raja S. C. Mullick Road Jadavpur, Kolkata 700032 West Bengal India), Fax
| |
Collapse
|
21
|
Cansiz S, Zhang L, Wu C, Wu Y, Teng IT, Hou W, Wang Y, Wan S, Cai R, Jin C, Liu Q, Tan W. DNA Aptamer Based Nanodrugs: Molecular Engineering for Efficiency. Chem Asian J 2015; 10:2084-94. [PMID: 26177853 DOI: 10.1002/asia.201500434] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Indexed: 12/21/2022]
Abstract
In the past two decades, the study of cancer therapy has gradually advanced to the "nano" era. Numerous novel nanomaterials armed with unique physical properties have been introduced into biomedical research. At the same time, functional nucleic acid molecules, especially aptamers, have aroused broad attention from the biomedical community. Benefiting from the advancement of molecular engineering strategies, it is now feasible to combine the cancer-specific recognition capability of aptamers with various other special functions of nanomaterials to develop cancer-specific drugs at the nanoscale. Nanodrugs are now offering an unprecedented opportunity to achieve the goal of efficient targeted delivery as well as controlled release. This review highlights some achievements made in multiple aptamer-based nanodrug systems that have emerged in recent years, including studies in the infant stage of "proof-of-concept".
Collapse
Affiliation(s)
- Sena Cansiz
- Departments of Chemistry, Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL, 32611-7200, USA
| | - Liqin Zhang
- Departments of Chemistry, Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL, 32611-7200, USA
| | - Cuichen Wu
- Departments of Chemistry, Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL, 32611-7200, USA.,Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering and Theranostics, Hunan University, Changsha, 410082, P.R. China
| | - Yuan Wu
- Departments of Chemistry, Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL, 32611-7200, USA.,Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering and Theranostics, Hunan University, Changsha, 410082, P.R. China
| | - I-Ting Teng
- Departments of Chemistry, Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL, 32611-7200, USA
| | - Weijia Hou
- Departments of Chemistry, Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL, 32611-7200, USA
| | - Yanyue Wang
- Departments of Chemistry, Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL, 32611-7200, USA
| | - Shuo Wan
- Departments of Chemistry, Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL, 32611-7200, USA
| | - Ren Cai
- Departments of Chemistry, Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL, 32611-7200, USA
| | - Chen Jin
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering and Theranostics, Hunan University, Changsha, 410082, P.R. China
| | - Qiaoling Liu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering and Theranostics, Hunan University, Changsha, 410082, P.R. China
| | - Weihong Tan
- Departments of Chemistry, Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL, 32611-7200, USA. .,Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Biology, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Molecular Engineering and Theranostics, Hunan University, Changsha, 410082, P.R. China.
| |
Collapse
|
22
|
Dinda S, Kakran M, Zeng J, Sudhaharan T, Ahmed S, Das D, Selvan ST. Grafting of ZnS:Mn-Doped Nanocrystals and an Anticancer Drug onto Graphene Oxide for Delivery and Cell Labeling. Chempluschem 2015; 81:100-107. [DOI: 10.1002/cplu.201500349] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Sanghamitra Dinda
- Institute of Materials Research and Engineering; Agency for Science, Technology and Research (A*STAR); 3 Research Link Singapore 117602 Singapore
- School of Pharmaceutical Sciences; Siksha O Anushandan University (SOA); Bhubaneshwar 751030 India
| | - Mitali Kakran
- Institute of Materials Research and Engineering; Agency for Science, Technology and Research (A*STAR); 3 Research Link Singapore 117602 Singapore
| | - Jialiu Zeng
- Institute of Materials Research and Engineering; Agency for Science, Technology and Research (A*STAR); 3 Research Link Singapore 117602 Singapore
- Department of Biomedical Engineering; Faculty of Engineering; National University of Singapore; 7 Engineering Drive 1 Singapore 117574 Singapore
| | - Thankiah Sudhaharan
- Neural Stem Cell Group; Institute of Medical Biology; 61 Biopolis Drive Singapore 138673 Republic of Singapore
| | - Sohail Ahmed
- Neural Stem Cell Group; Institute of Medical Biology; 61 Biopolis Drive Singapore 138673 Republic of Singapore
| | - Debajyoti Das
- School of Pharmaceutical Sciences; Siksha O Anushandan University (SOA); Bhubaneshwar 751030 India
| | - Subramanian Tamil Selvan
- Institute of Materials Research and Engineering; Agency for Science, Technology and Research (A*STAR); 3 Research Link Singapore 117602 Singapore
- Department of Biomedical Engineering; Faculty of Engineering; National University of Singapore; 7 Engineering Drive 1 Singapore 117574 Singapore
| |
Collapse
|
23
|
Whitfield CJ, Turley AT, Tuite EM, Connolly BA, Pike AR. Enzymatic Method for the Synthesis of Long DNA Sequences with Multiple Repeat Units. Angew Chem Int Ed Engl 2015; 54:8971-4. [DOI: 10.1002/anie.201502971] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Indexed: 11/09/2022]
|
24
|
Whitfield CJ, Turley AT, Tuite EM, Connolly BA, Pike AR. Enzymatic Method for the Synthesis of Long DNA Sequences with Multiple Repeat Units. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201502971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
25
|
Zhao TT, Chen QY, Yang H. Spectroscopic study on the formation of DNA-Ag clusters and its application in temperature sensitive vehicles of DOX. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2015; 137:66-69. [PMID: 25200118 DOI: 10.1016/j.saa.2014.08.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 06/27/2014] [Accepted: 08/21/2014] [Indexed: 06/03/2023]
Abstract
DNA silver nanoclusters (DNA-AgNCs) with a fluorescence emission at 610 nm were synthesized using a special hairpin DNA sequence (5'-AGCACGTAG-C3AC3AC3GC3A-CTACGTGCT-3'). Spectroscopic data demonstrate that the DNA changed from an i-motif structure containing C-quadruplexes to anti-parallel four strands structure during the formation of DNA-AgNCs. Importantly, the loose and compact four strand structure caused by the melting and hybridization of stem duplex was confirmed by the reversible fluorescence change of DNA-AgNCs in the range of 25-66°C. Herein, DNA-AgNCs were used as temperature sensitive vehicles of drug loading. The drug loading capacity is 1 Doxorubicin (Dox) molecules per CG pairs on stem-duplexes. The loaded Dox can be released by raising temperature with the melt of stem duplex. Moreover, the special DNA sequence makes it sensitive to the HepG-2 cells.
Collapse
Affiliation(s)
- Ting-Ting Zhao
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Qiu-Yun Chen
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, PR China.
| | - Huan Yang
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, PR China
| |
Collapse
|
26
|
Kruspe S, Mittelberger F, Szameit K, Hahn U. Aptamers as drug delivery vehicles. ChemMedChem 2014; 9:1998-2011. [PMID: 25130604 DOI: 10.1002/cmdc.201402163] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/02/2014] [Indexed: 01/22/2023]
Abstract
The benefits of directed and selective therapy for systemic treatment are reasons for increased interest in exploiting aptamers for cell-specific drug delivery. Nucleic acid based pharmaceuticals represent an interesting and novel tool to counter human diseases. Combining inhibitory potential and cargo transfer upon internalization, nanocarriers as well as various therapeutics including siRNAs, chemotherapeutics, photosensitizers, or proteins can be imported via these synthetic nucleic acids. However, widespread clinical application is still hampered by obstacles that must be overcome. In this review, we give an overview of applications and recent advances in aptamer-mediated drug delivery. We also introduce prominent selection methods as well as useful approaches in choice of drug and conjugation method. We discuss the challenges that need to be considered and present strategies that have been applied to achieve intracellular delivery of effectors transported by readily internalized aptamers.
Collapse
Affiliation(s)
- Sven Kruspe
- Institut für Biochemie und Molekularbiologie, Universität Hamburg, Martin-Luther-King Platz 6, 20146 Hamburg (Germany)
| | | | | | | |
Collapse
|
27
|
Mo R, Jiang T, Gu Z. Enhanced anticancer efficacy by ATP-mediated liposomal drug delivery. Angew Chem Int Ed Engl 2014; 53:5815-20. [PMID: 24764317 DOI: 10.1002/anie.201400268] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Indexed: 11/12/2022]
Abstract
A liposome-based co-delivery system composed of a fusogenic liposome encapsulating ATP-responsive elements with chemotherapeutics and a liposome containing ATP was developed for ATP-mediated drug release triggered by liposomal fusion. The fusogenic liposome had a protein-DNA complex core containing an ATP-responsive DNA scaffold with doxorubicin (DOX) and could release DOX through a conformational change from the duplex to the aptamer/ATP complex in the presence of ATP. A cell-penetrating peptide-modified fusogenic liposomal membrane was coated on the core, which had an acid-triggered fusogenic potential with the ATP-loaded liposomes or endosomes/lysosomes. Directly delivering extrinsic liposomal ATP promoted the drug release from the fusogenic liposome in the acidic intracellular compartments upon a pH-sensitive membrane fusion and anticancer efficacy was enhanced both in vitro and in vivo.
Collapse
Affiliation(s)
- Ran Mo
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695 (USA); Molecular Pharmaceutics Division, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 (USA).
| | | | | |
Collapse
|
28
|
Mo R, Jiang T, Gu Z. Enhanced Anticancer Efficacy by ATP-Mediated Liposomal Drug Delivery. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201400268] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
29
|
Zhang Y, Yin Q, Yin L, Ma L, Tang L, Cheng J. Chain-shattering polymeric therapeutics with on-demand drug-release capability. Angew Chem Int Ed Engl 2013; 52:6435-9. [PMID: 23650111 PMCID: PMC3800742 DOI: 10.1002/anie.201300497] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Revised: 02/19/2013] [Indexed: 11/07/2022]
Abstract
Design of smart polymeric therapeutics We designed and synthesized trigger-responsive chain-shattering polymeric therapeutics (CSPTs) via condensation polymerization of a UV-or hydrogen peroxide-responsive domain and a bisfunctional drug as co-monomers. CSPTs have precisely controlled molecular composition and unique chain-shattering type of drug release mechanism. Drug release kinetics can be precisely controlled by means of the trigger treatment. Chemotherapeutic-containing CSPTs showed trigger-responsive in vitro and in vivo antitumor efficacy.
Collapse
Affiliation(s)
- Yanfeng Zhang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, 1304 West Green Street, Urbana, IL, 61801 (USA), Fax: (+1) 217-333-2736
| | - Qian Yin
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, 1304 West Green Street, Urbana, IL, 61801 (USA), Fax: (+1) 217-333-2736
| | - Lichen Yin
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, 1304 West Green Street, Urbana, IL, 61801 (USA), Fax: (+1) 217-333-2736
| | - Liang Ma
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, 1304 West Green Street, Urbana, IL, 61801 (USA), Fax: (+1) 217-333-2736
| | - Li Tang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, 1304 West Green Street, Urbana, IL, 61801 (USA), Fax: (+1) 217-333-2736
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, 1304 West Green Street, Urbana, IL, 61801 (USA), Fax: (+1) 217-333-2736
| |
Collapse
|
30
|
Zhang Y, Yin Q, Yin L, Ma L, Tang L, Cheng J. Chain-Shattering Polymeric Therapeutics with On-Demand Drug-Release Capability. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201300497] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
31
|
Lv S, Guan Y, Wang D, Du Y. Aptamer based strategy for cytosensing and evaluation of HER-3 on the surface of MCF-7 cells by using the signal amplification of nucleic acid-functionalized nanocrystals. Anal Chim Acta 2013; 772:26-32. [PMID: 23540244 DOI: 10.1016/j.aca.2013.02.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Revised: 02/05/2013] [Accepted: 02/11/2013] [Indexed: 10/27/2022]
Abstract
The electrochemical detection of cell lines of MCF-7 (human breast cancer) has been reported, using magnetic beads for the separation tool and high-affinity DNA aptamers for signal recognition. The high specificity was obtained by using the magnetic beads and aptamers, and the good sensitivity was realized with the signal amplification of DNA capped CdS or PbS nanocrystals. The ASV (anodic stripping voltammetry) technology was employed for the detection of cadmic cation and lead ions, for electrochemical assay of the amount of the target cells and biomarkers on the membrane of target cells, respectively. This electrochemical method could respond to as low as 100 cells mL(-1) of cancer cells with a linear calibration range from 1.0×10(2) to 1.0×10(6) cells mL(-1), showing very high sensitivity. Moreover, the amounts of HER-3 which were overexpressed on MCF-7 cells were calculated correspond to be 3.56×10(4) anti-HER-3 antibody molecules. In addition, the assay was able to differentiate between different types of target and control cells based on the aptamers and magnetic beads used in the assay, indicating the wide applicability of the assay for early and accurate diagnose of cancers.
Collapse
Affiliation(s)
- Shaoping Lv
- Departmen of Neurology, Provinvial Hospital affiliated Shandong University, Jinan, Shandong, PR China
| | | | | | | |
Collapse
|
32
|
Xiao Z, Ji C, Shi J, Pridgen EM, Frieder J, Wu J, Farokhzad OC. DNA Self-Assembly of Targeted Near-Infrared-Responsive Gold Nanoparticles for Cancer Thermo-Chemotherapy. Angew Chem Int Ed Engl 2012. [DOI: 10.1002/ange.201204018] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
33
|
Xiao Z, Ji C, Shi J, Pridgen EM, Frieder J, Wu J, Farokhzad OC. DNA self-assembly of targeted near-infrared-responsive gold nanoparticles for cancer thermo-chemotherapy. Angew Chem Int Ed Engl 2012; 51:11853-7. [PMID: 23081716 PMCID: PMC3532659 DOI: 10.1002/anie.201204018] [Citation(s) in RCA: 248] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 09/18/2012] [Indexed: 11/08/2022]
Affiliation(s)
- Zeyu Xiao
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Zhu G, Meng L, Ye M, Yang L, Sefah K, O’Donoghue MB, Chen Y, Xiong X, Huang J, Song E, Tan W. Self-assembled aptamer-based drug carriers for bispecific cytotoxicity to cancer cells. Chem Asian J 2012; 7:1630-6. [PMID: 22492537 PMCID: PMC3475610 DOI: 10.1002/asia.201101060] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Indexed: 11/10/2022]
Abstract
Monovalent aptamers can deliver drugs to target cells by specific recognition. However, different cancer subtypes are distinguished by heterogeneous biomarkers and one single aptamer is unable to recognize all clinical samples from different patients with even the same type of cancers. To address heterogeneity among cancer subtypes for targeted drug delivery, as a model, we developed a drug carrier with a broader recognition range of cancer subtypes. This carrier, sgc8c-sgd5a (SD), was self-assembled from two modified monovalent aptamers. It showed bispecific recognition abilities to target cells in cell mixtures; thus broadening the recognition capabilities of its parent aptamers. The self-assembly of SD simultaneously formed multiple drug loading sites for the anticancer drug doxorubicin (Dox). The Dox-loaded SD (SD-Dox) also showed bispecific abilities for target cell binding and drug delivery. Most importantly, SD-Dox induced bispecific cytotoxicity in target cells in cell mixtures. Therefore, by broadening the otherwise limited recognition capabilities of monovalent aptamers, bispecific aptamer-based drug carriers would facilitate aptamer applications for clinically heterogeneous cancer subtypes that respond to the same cancer therapy.
Collapse
Affiliation(s)
- Guizhi Zhu
- Center for Research at the Bio/Nano Interface, Departments of Chemistry, Physiology and Functional Genomics, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200 (USA), Fax: (+ 1) 352-846-2410
| | - Ling Meng
- Center for Research at the Bio/Nano Interface, Departments of Chemistry, Physiology and Functional Genomics, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200 (USA), Fax: (+ 1) 352-846-2410
| | - Mao Ye
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology and College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, People’s Republic of China
| | - Liu Yang
- Center for Research at the Bio/Nano Interface, Departments of Chemistry, Physiology and Functional Genomics, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200 (USA), Fax: (+ 1) 352-846-2410
| | - Kwame Sefah
- Center for Research at the Bio/Nano Interface, Departments of Chemistry, Physiology and Functional Genomics, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200 (USA), Fax: (+ 1) 352-846-2410
| | - Meghan B. O’Donoghue
- Center for Research at the Bio/Nano Interface, Departments of Chemistry, Physiology and Functional Genomics, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200 (USA), Fax: (+ 1) 352-846-2410
| | - Yan Chen
- Center for Research at the Bio/Nano Interface, Departments of Chemistry, Physiology and Functional Genomics, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200 (USA), Fax: (+ 1) 352-846-2410
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology and College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, People’s Republic of China
| | - Xiangling Xiong
- Center for Research at the Bio/Nano Interface, Departments of Chemistry, Physiology and Functional Genomics, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200 (USA), Fax: (+ 1) 352-846-2410
| | - Jin Huang
- Center for Research at the Bio/Nano Interface, Departments of Chemistry, Physiology and Functional Genomics, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200 (USA), Fax: (+ 1) 352-846-2410
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Biology and College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, People’s Republic of China
| | - Erqun Song
- Center for Research at the Bio/Nano Interface, Departments of Chemistry, Physiology and Functional Genomics, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200 (USA), Fax: (+ 1) 352-846-2410
- Key Laboratory of Luminescence and Real-Time Analysis of the Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Weihong Tan
- Center for Research at the Bio/Nano Interface, Departments of Chemistry, Physiology and Functional Genomics, Shands Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200 (USA), Fax: (+ 1) 352-846-2410
| |
Collapse
|
35
|
Tan L, Neoh KG, Kang ET, Choe WS, Su X. PEGylated anti-MUC1 aptamer-doxorubicin complex for targeted drug delivery to MCF7 breast cancer cells. Macromol Biosci 2011; 11:1331-5. [PMID: 21818856 DOI: 10.1002/mabi.201100173] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2011] [Revised: 07/02/2011] [Indexed: 12/12/2022]
Abstract
Targeted drug delivery is especially important in cancer treatment as many anti-cancer drugs are non-specific and highly toxic to both cancer and normal cells. The targeted drug delivery of DOX to the MUC1-expressing breast cancer cell line (MCF7) was obtained using APT as a carrier. Modification of the APT-DOX complex by PEG increases the survivability of the macrophage control (RAW 264.7) by about six-fold as compared to free DOX treatment without significantly affecting the cytotoxicity toward the target cell line. Thus, PEG-APT-DOX is potentially a new therapeutic agent for targeted drug delivery to MUC1-expressing cell lines.
Collapse
Affiliation(s)
- Lihan Tan
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Kent Ridge, 119260, Singapore
| | | | | | | | | |
Collapse
|
36
|
|
37
|
|
38
|
Self-organized Nanogels of Polysaccharide Derivatives in Anti-Cancer Drug Delivery. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2010. [DOI: 10.4333/kps.2010.40.4.201] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
39
|
Tang Z, Zhu Z, Mallikaratchy P, Yang R, Sefah K, Tan W. Aptamer-target binding triggered molecular mediation of singlet oxygen generation. Chem Asian J 2010; 5:783-6. [PMID: 20340154 DOI: 10.1002/asia.200900545] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Zhiwen Tang
- Department of Chemistry, Shands Cancer Center and UF Genetics Institute, University of Florida, Gainesville, Florida 32611-7200, USA
| | | | | | | | | | | |
Collapse
|
40
|
Kim E, Jung Y, Choi H, Yang J, Suh JS, Huh YM, Kim K, Haam S. Prostate cancer cell death produced by the co-delivery of Bcl-xL shRNA and doxorubicin using an aptamer-conjugated polyplex. Biomaterials 2010; 31:4592-9. [PMID: 20206379 DOI: 10.1016/j.biomaterials.2010.02.030] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 02/10/2010] [Indexed: 11/19/2022]
Abstract
We investigated the synergism between shRNAs against Bcl-xL and doxorubicin (DOX) using aptamer-conjugated polyplexes (APs) in combination cancer therapy. Synergistic and selective cancer cell death was achieved by AP-mediated co-delivery of very small amounts of DOX and Bcl-xL-specific shRNA, which simultaneously activated an intrinsic apoptotic pathway. A branched polyethyleneimine (PEI) was grafted to polyethylene glycol (PEI-PEG) to serve as a vehicle for shRNA delivery, and its surface was further conjugated with an anti-PSMA aptamer (APT) for the selective delivery of APs to prostate cancer cells that express prostate-specific membrane antigens (PSMA) on their cell surface. The APs were finally obtained after intercalation of DOX to form shRNA/PEI-PEG-APT/DOX conjugates. Cell viability assays and FACS analysis of GFP expression against PC3 (PSMA deficient) and LNCaP (PSMA overexpressed) cells demonstrated that the synthesized APs inhibited the growth of PSMA-abundant prostate cancer cells with strong cell selectivity. Consequently, IC(50) values of APs loaded with both DOX and shRNA were approximately 17-fold less than those for the simple mixture of shRNA plus drug (shRNA/Lipofectamine + DOX). These results suggest that AP-mediated co-delivery of an anti-cancer drug and shRNA against Bcl-xL may widen the therapeutic window and allow for the selective destruction of cancer cells.
Collapse
Affiliation(s)
- Eunjung Kim
- Department of Chemical and Biomolecular Engineering, College of Engineering, Yonsei University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Huang YF, Shangguan D, Liu H, Phillips JA, Zhang X, Chen Y, Tan W. Molecular assembly of an aptamer-drug conjugate for targeted drug delivery to tumor cells. Chembiochem 2009; 10:862-8. [PMID: 19253922 DOI: 10.1002/cbic.200800805] [Citation(s) in RCA: 286] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The conjugation of antitumor drugs to targeting reagents such as antibodies is a promising method that can increase the efficacy of chemotherapy and reduce the overall toxicity of the drugs. In this study, we covalently link the antitumor agent doxorubicin (Dox) to the DNA aptamer sgc8c, which was selected by the cell-SELEX method. In doing so, we expected that this sgc8c-Dox conjugate would specifically kill the target CCRF-CEM (T-cell acute lymphoblastic leukemia, T-cell ALL) cells, but with minimal toxicity towards nontarget cells. The results demonstrated that the sgc8c-Dox conjugate possesses many of the properties of the sgc8c aptamer, including high binding affinity (K(d)=2.0+/-0.2 nM) and the capability to be efficiently internalized by target cells. Moreover, due to the specific conjugation method, the acid-labile linkage connecting the sgc8c-Dox conjugate can be cleaved inside the acidic endosomal environment. Cell viability tests demonstrate that the sgc8c-Dox conjugates not only possess potency similar to unconjugated Dox, but also have the required molecular specificity that is lacking in most current targeted drug delivery strategies. Furthermore, we found that nonspecific uptake of membrane-permeable Dox to nontarget cell lines could also be inhibited by linking the drug with the aptamer; thus, the conjugates are selective for cells that express higher amounts of target proteins. Compared to the less effective Dox-immunoconjugates, these sgc8c-Dox conjugates make targeted chemotherapy more feasible with drugs having various potencies. When combined with the large number of recently created DNA aptamers that specifically target a wide variety of cancer cells, this drug-aptoconjugation method will have broad implications for targeted drug delivery.
Collapse
Affiliation(s)
- Yu-Fen Huang
- Department of Chemistry, Shands Cancer Center and Center for Research at Bio/Nano Interface, Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Aptamers are small single-stranded nucleic acids that fold into a well-defined three-dimensional structure. They show a high affinity and specificity for their target molecules and inhibit their biological functions. Aptamers belong to the nucleic acids family and can be synthesized by chemical or enzymatic procedures, or a combination of the two. They can, therefore, be considered as both chemical and biological substances. This Review summarizes the most convenient approaches to their preparation and new developments in the field of aptamers. The application of aptamers in chemical biology is also discussed.
Collapse
Affiliation(s)
- Günter Mayer
- Life and Medical Sciences, Prog. Unit Chemical Biology and Medicinal Chemistry, University of Bonn c/o Kekulé-Institute for Organic Chemistry and Biochemistry, Gerhard-Domagk-Strasse 1, 53121 Bonn, Germany.
| |
Collapse
|
43
|
|
44
|
Zhang L, Radovic-Moreno AF, Alexis F, Gu FX, Basto PA, Bagalkot V, Jon S, Langer RS, Farokhzad OC. Co-delivery of hydrophobic and hydrophilic drugs from nanoparticle-aptamer bioconjugates. ChemMedChem 2008; 2:1268-71. [PMID: 17600796 DOI: 10.1002/cmdc.200700121] [Citation(s) in RCA: 177] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Liangfang Zhang
- Department of Chemical Engineering and Division of Health Science and Technology, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Biomaterialien: Langer und Fratzl ausgezeichnet / Organische Chemie: Itami geehrt. Angew Chem Int Ed Engl 2008. [DOI: 10.1002/ange.200802771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
46
|
Biomaterials: Awards to Langer and Fratzl / Organic Chemistry: Itami Honored. Angew Chem Int Ed Engl 2008. [DOI: 10.1002/anie.200802771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
47
|
Yu M, Jeong Y, Park J, Park S, Kim J, Min J, Kim K, Jon S. Drug‐Loaded Superparamagnetic Iron Oxide Nanoparticles for Combined Cancer Imaging and Therapy In Vivo. Angew Chem Int Ed Engl 2008. [DOI: 10.1002/ange.200800857] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
48
|
Yu M, Jeong Y, Park J, Park S, Kim J, Min J, Kim K, Jon S. Drug‐Loaded Superparamagnetic Iron Oxide Nanoparticles for Combined Cancer Imaging and Therapy In Vivo. Angew Chem Int Ed Engl 2008; 47:5362-5. [DOI: 10.1002/anie.200800857] [Citation(s) in RCA: 503] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
49
|
Li W, Yang X, Wang K, Tan W, He Y, Guo Q, Tang H, Liu J. Real-time imaging of protein internalization using aptamer conjugates. Anal Chem 2008; 80:5002-8. [PMID: 18533682 DOI: 10.1021/ac800930q] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Angiogenin is a potent angiogenic factor that is known to play an important role in tumor angiogenesis. In this paper, we investigate the cellular internalization of angiogenin conjugated with its highly specific aptamer. By using fluorophore-labeled aptamer and confocal laser scanning microscopy, we have developed a novel and simple method by which to visualize the real-time process of angiogenin internalization. Specifically, when aptamer-angiogenin conjugates were added into cell cultures, conjugates could be selectively bound to HUVE cells (human umbilical vein endothelial cells) and MCF-7 cells (human breast cancer cells). Nuclear staining and Z-axis scanning studies demonstrated that the aptamer-angiogenin conjugates were internalized to intracellular organelles, and dynamic confocal imaging studies indicated that the conjugates were quickly internalized. These results provide the first evidence that a fluorophore-labeled aptamer can be used as a fluorescent probe to visualize the spatiotemporal process of protein internalization in real time.
Collapse
Affiliation(s)
- Wei Li
- College of Chemistry and Chemical Engineering, Engineering Center for Biomedicine, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, PR China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Xiao Z, Shangguan D, Cao Z, Fang X, Tan W. Cell-specific internalization study of an aptamer from whole cell selection. Chemistry 2008; 14:1769-75. [PMID: 18092308 DOI: 10.1002/chem.200701330] [Citation(s) in RCA: 190] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Nucleic acid aptamers have been shown many unique applications as excellent probes in molecular recognition. However, few examples are reported which show that aptamers can be internalized inside living cells for aptamer functional studies and for targeted intracellular delivery. This is mainly due to the limited number of aptamers available for cell-specific recognition, and the lack of research on their extra- and intracellular functions. One of the major difficulties in aptamers' in vivo application is that most of aptamers, unlike small molecules, cannot be directly taken up by cells without external assistance. In this work, we have studied a newly developed and cell-specific DNA aptamer, sgc8. This aptamer has been selected through a novel cell selection process (cell-SELEX), in which whole intact cells are used as targets while another related cell line is used as a negative control. The cell-SELEX enables generation of multiple aptamers for molecular recognition of the target cells and has significant advantages in discovering cell surface binding molecules for the selected aptamers. We have studied the cellular internalization of one of the selected aptamers. Our results show that sgc8 is internalized efficiently and specifically to the lymphoblastic leukemia cells. The internalized sgc8 aptamers are located inside the endosome. Comparison studies are done with the antibody for the binding protein of sgc8, PTK7 (Human protein tyrosine kinase-7) on cell surface. We also studied the internalization kinetics of both the aptamer and the antibody for the same protein on the living cell surface. We have further evaluated the effects of sgc8 on cell viability, and no cytotoxicity is observed. This study indicates that sgc8 is a promising agent for cell-type specific intracellular delivery.
Collapse
Affiliation(s)
- Zeyu Xiao
- Center for Research at the Bio/Nano Interface, Department of Chemistry, Shands Cancer Center and UF Genetics Institute, McKnight Brain Institute, University of Florida, Gainesville, FL 32611-7200, USA
| | | | | | | | | |
Collapse
|