1
|
Coppola G, Cennamo F, Ciccone G, Ibba ML, Ruscio AD, Vito AD, Esposito CL, Catuogno S. Aptamer-based applications in delivering cancer gene therapies and beyond: state of the art and the missing links to clinical translation. Adv Drug Deliv Rev 2025:115639. [PMID: 40513886 DOI: 10.1016/j.addr.2025.115639] [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: 04/11/2025] [Revised: 06/03/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
The possibility of correcting genetic and epigenetic alterations through gene therapies has been considered a cornerstone in oncology. However, modest results have been achieved in clinics, mainly due to inefficient tumor targeting and side effects. Nucleic acid aptamers are three-dimensional folded single-stranded DNAs or RNAs that selectively bind receptors on cellular membranes, being subsequently internalized via receptor-mediated endocytosis. Thanks to this capability, internalizing aptamers have been investigated as targeting moieties to deliver gene therapies more efficiently and selectively in tumor cells. Promising preclinical results suggested that aptamers could represent the long-awaited step forward in cancer gene therapy. Nevertheless, no clinical trials of aptamer-based gene therapies have been carried out two decades after the first preclinical application, indicating the field could not be sufficiently mature for translatability. The review aims to update thestate of the art regarding aptamers' contribution to gene therapy delivery and to critically highlight the main shortcomings that could have hindered clinical evaluations. In addition, pioneering insights regarding the use of aptamers as co-factors in CRISPR/Cas9 technology or as direct epigenetic regulators are also summarized, revealing more extended applicability not limited to the delivery of cancer gene therapies.
Collapse
Affiliation(s)
- Gabriele Coppola
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| | - Fabiola Cennamo
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| | - Giuseppe Ciccone
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy; Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", 81100 Caserta, Italy.
| | - Maria Luigia Ibba
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy; Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Caserta, Italy.
| | - Annalisa Di Ruscio
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA; Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Boston, MA 02215, USA.
| | - Aldo Di Vito
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| | - Carla Lucia Esposito
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| | - Silvia Catuogno
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) of the Italian National Research Council (CNR), Naples, Italy.
| |
Collapse
|
2
|
Xia B, Zhu Q. Aptamer-ODN Chimeras: Enabling Cell-Specific ODN Targeting Therapy. Cells 2025; 14:697. [PMID: 40422200 DOI: 10.3390/cells14100697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/23/2025] [Accepted: 05/10/2025] [Indexed: 05/28/2025] Open
Abstract
Oligonucleotides (ODNs) such as siRNA, saRNA, and miRNA regulate gene expression through a variety of molecular mechanisms and show unique potential in the treatment of genetic diseases and rare diseases, but their clinical application is still limited by the efficiency of the delivery system, especially the problem of the insufficient targeting of extrahepatic tissues. As homologous nucleic acid molecules, aptamers have become a key tool to improve the targeted delivery of ODNs. Aptamer-ODN chimeras can not only bind to multiple proteins on the cell surface with high specificity and selectivity, but they can also internalize into cells. Furthermore, they outperform traditional delivery systems in terms of cost-effectiveness and chemical modification flexibility. This review systematically summarizes the origin and progress of aptamer-ODN chimera therapy, discusses some innovative design strategies, and proposes views on the future direction of aptamer-ODN chimeras.
Collapse
Affiliation(s)
- Bei Xia
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Qubo Zhu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| |
Collapse
|
3
|
Xue L, Yu X, Zhao L, Garrett A, Wu D, Liu HY. Targeted Delivery of AR-V7 siRNA with Bivalent PSMA Aptamers Effectively Suppresses the Growth of Enzalutamide-Resistant Prostate Cancer. Mol Pharm 2024; 21:5749-5760. [PMID: 39388218 DOI: 10.1021/acs.molpharmaceut.4c00743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Androgen deprivation therapy has been the primary treatment strategy for advanced prostate cancer (PCa). But most patients develop castration resistance over time. For FDA-approved second-generation androgen receptor (AR) antagonists, including enzalutamide (ENZ) and abiraterone (AA), patients who initially respond to them eventually develop resistance. The key mechanism for resistance to ENZ/AA involves AR splice variants (AR-Vs) and specifically AR-V7. Current AR antagonists cannot target AR-V7 due to its lack of the C-terminal ligand-binding domain (LBD) but keeping the AR N-terminal domain (NTD) which still can activate androgen-responsive genes. Therefore, targeting the AR NTD and AR-V7 is critically important to overcome ENZ resistance. Unfortunately, AR NTD has been considered an "undruggable" target due to the difficulty in defining its three-dimensional (3D) structure. In this context, siRNA is highly suitable to address this undruggable target. However, siRNA cannot freely diffuse into cells, and a carrier is needed. In this regard, nucleic acid-based aptamers are highly suitable for cell type-specific delivery of siRNA in vivo. In this study, we have developed a serum-stable bivalent prostate-specific membrane antigen (PSMA) aptamer-AR-V7 siRNA chimera (PAP). The results show that PAP can knock down both AR-full length and AR-V7 in PSMA-expressing castration-resistant cells. It can resensitize ENZ in cell lines and PCa xenografts. ENZ combined with PAP can significantly inhibit 22Rv1 xenograft growth in mice without experiencing castration. Owing to the low toxicity, PAP has potential to offer a new antiandrogen treatment for current ENZ-resistant PCa.
Collapse
MESH Headings
- Male
- Phenylthiohydantoin
- Humans
- Animals
- Benzamides
- Nitriles
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Mice
- RNA, Small Interfering/administration & dosage
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Glutamate Carboxypeptidase II/antagonists & inhibitors
- Glutamate Carboxypeptidase II/genetics
- Glutamate Carboxypeptidase II/metabolism
- Xenograft Model Antitumor Assays
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/pathology
- Aptamers, Nucleotide
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Mice, Nude
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/pathology
- Androgen Receptor Antagonists/pharmacology
- Cell Proliferation/drug effects
Collapse
Affiliation(s)
- Lu Xue
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912, United States
- Department of Pediatrics Hematology, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaolin Yu
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912, United States
| | - Lijing Zhao
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912, United States
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun 130021, China
| | - Aria Garrett
- Dotquant, CoMotion at University of Washington, Seattle, Washington 98195, United States
| | - Daqing Wu
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912, United States
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, Georgia 30314, United States
| | - Hong Yan Liu
- Georgia Cancer Center, Augusta University, Augusta, Georgia 30912, United States
- Dotquant, CoMotion at University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
4
|
Zhang T, Yin K, Niu X, Bai X, Wang Z, Ji M, Yuan B. Development of Bivalent Aptamer-DNA Carrier-Doxorubicin Conjugates for Targeted Killing of Esophageal Squamous Cell Carcinoma Cells. Int J Mol Sci 2024; 25:7959. [PMID: 39063201 PMCID: PMC11276760 DOI: 10.3390/ijms25147959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Esophageal cancer ranks the seventh in cancer incidence and the sixth in cancer death. Esophageal squamous cell carcinoma (ESCC) accounts for approximately 90% of the total cases of esophageal cancer. Chemotherapy is the most effective drug-based method for treatment of esophageal cancer. However, severe side effects of traditional chemotherapy limit its treatment efficacy. Targeted chemotherapy can deliver chemotherapeutic drugs to cancer cells and specifically kill these cells with reduced side effects. In the work, the bivalent aptamer-DNA carrier (BAD) was designed by using an ESCC cell-specific aptamer as the recognition molecule and a GC base-rich DNA sequence as the drug carrier. With doxorubicin (Dox) as chemotherapeutic drugs, the bivalent aptamer-DNA-Dox conjugate (BADD) was constructed for targeted killing of ESCC cells. Firstly, the truncated A2(35) aptamer with a retained binding ability was obtained through optimization of an intact A2(80) aptamer and was used to fuse with DNA carrier sequences for constructing the BAD through simple DNA hybridization. The results of gel electrophoresis and flow cytometry analysis showed that the BAD was successfully constructed and had a stronger binding affinity than monovalent A2(35). Then, the BAD was loaded with Dox drugs to construct the BADD through noncovalent intercalation. The results of fluorescence spectra and flow cytometry assays showed that the BADD was successfully constructed and can bind to target cells strongly. Confocal imaging further displayed that the BADD can be specifically internalized into target cells and release Dox. The results of CCK-8 assays, Calcein AM/PI staining, and wound healing assays demonstrated that the BADD can specifically kill target cells, but not control cells. Our results demonstrate that the developed BADD can specifically deliver doxorubicin to target ESCC cells and selectively kill these cells, offering a potentially effective strategy for targeted chemotherapy of ESCC.
Collapse
Affiliation(s)
- Tianlu Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
| | - Kai Yin
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
| | - Xidong Niu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
| | - Xue Bai
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
| | - Zhaoting Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
| | - Mengmeng Ji
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
| | - Baoyin Yuan
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; (T.Z.); (K.Y.); (X.N.); (X.B.); (Z.W.); (M.J.)
- Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
5
|
Singh S, Srivastava P. Targeted Protein Degraders- The Druggability Perspective. J Pharm Sci 2024; 113:539-554. [PMID: 37926234 DOI: 10.1016/j.xphs.2023.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/14/2023] [Accepted: 10/14/2023] [Indexed: 11/07/2023]
Abstract
Targeted Protein degraders (TPDs) show promise in harnessing cellular machinery to eliminate disease-causing proteins, even those previously considered undruggable. Especially if protein turnover is low, targeted protein removal bestows lasting therapeutic effect over typical inhibition. The demonstrated safety and efficacy profile of clinical candidates has fueled the surge in the number of potential candidates across different therapeutic areas. As TPDs often do not comply with Lipinski's rule of five, developing novel TPDs and unlocking their full potential requires overcoming solubility, permeability and oral bioavailability challenges. Tailored in-vitro assays are key to precise profiling and optimization, propelling breakthroughs in targeted protein degradation.
Collapse
|
6
|
Shishparenok AN, Furman VV, Zhdanov DD. DNA-Based Nanomaterials as Drug Delivery Platforms for Increasing the Effect of Drugs in Tumors. Cancers (Basel) 2023; 15:2151. [PMID: 37046816 PMCID: PMC10093432 DOI: 10.3390/cancers15072151] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023] Open
Abstract
DNA nanotechnology has significantly advanced and might be used in biomedical applications, drug delivery, and cancer treatment during the past few decades. DNA nanomaterials are widely used in biomedical research involving biosensing, bioimaging, and drug delivery since they are remarkably addressable and biocompatible. Gradually, modified nucleic acids have begun to be employed to construct multifunctional DNA nanostructures with a variety of architectural designs. Aptamers are single-stranded nucleic acids (both DNAs and RNAs) capable of self-pairing to acquire secondary structure and of specifically binding with the target. Diagnosis and tumor therapy are prospective fields in which aptamers can be applied. Many DNA nanomaterials with three-dimensional structures have been studied as drug delivery systems for different anticancer medications or gene therapy agents. Different chemical alterations can be employed to construct a wide range of modified DNA nanostructures. Chemically altered DNA-based nanomaterials are useful for drug delivery because of their improved stability and inclusion of functional groups. In this work, the most common oligonucleotide nanomaterials were reviewed as modern drug delivery systems in tumor cells.
Collapse
Affiliation(s)
- Anastasiya N. Shishparenok
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia
| | - Vitalina V. Furman
- Center of Chemical Engineering, ITMO University, Kronverkskiy Prospekt 49A, 197101 St. Petersburg, Russia
| | - Dmitry D. Zhdanov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya St. 6, 117198 Moscow, Russia
| |
Collapse
|
7
|
He S, Du Y, Tao H, Duan H. Advances in aptamer-mediated targeted delivery system for cancer treatment. Int J Biol Macromol 2023; 238:124173. [PMID: 36965552 DOI: 10.1016/j.ijbiomac.2023.124173] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
Aptamers with high affinity and specificity for certain targets have rapidly become a novel class of targeted ligands applicated in drug delivery. Based on the excellent characteristics of aptamers, different aptamer-mediated drug delivery systems have been developed, including aptamer-drug conjugate (ApDC), aptamer-siRNA, and aptamer-functionalized nanoparticle systems for the effective treatment of cancer, which can reduce potential toxicity and improve therapeutic efficacy. In this review, we summarize the recent progress of aptamer-mediated delivery systems in cancer therapy, and discuss the application prospects and existing problems of innovative approaches based on aptamer therapy. Overall, this review aims to better understand the current aptamer-based targeted delivery applications through in-depth analysis to improve efficacy and develop new therapeutic methods which can ultimately improve treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- Shiming He
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongyu Tao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Huaiyu Duan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
8
|
Gorachinov F, Mraiche F, Moustafa DA, Hishari O, Ismail Y, Joseph J, Crcarevska MS, Dodov MG, Geskovski N, Goracinova K. Nanotechnology - a robust tool for fighting the challenges of drug resistance in non-small cell lung cancer. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2023; 14:240-261. [PMID: 36865093 PMCID: PMC9972888 DOI: 10.3762/bjnano.14.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
Genomic and proteomic mutation analysis is the standard of care for selecting candidates for therapies with tyrosine kinase inhibitors against the human epidermal growth factor receptor (EGFR TKI therapies) and further monitoring cancer treatment efficacy and cancer development. Acquired resistance due to various genetic aberrations is an unavoidable problem during EGFR TKI therapy, leading to the rapid exhaustion of standard molecularly targeted therapeutic options against mutant variants. Attacking multiple molecular targets within one or several signaling pathways by co-delivery of multiple agents is a viable strategy for overcoming and preventing resistance to EGFR TKIs. However, because of the difference in pharmacokinetics among agents, combined therapies may not effectively reach their targets. The obstacles regarding the simultaneous co-delivery of therapeutic agents at the site of action can be overcome using nanomedicine as a platform and nanotools as delivery agents. Precision oncology research to identify targetable biomarkers and optimize tumor homing agents, hand in hand with designing multifunctional and multistage nanocarriers that respond to the inherent heterogeneity of the tumors, may resolve the challenges of inadequate tumor localization, improve intracellular internalization, and bring advantages over conventional nanocarriers.
Collapse
Affiliation(s)
- Filip Gorachinov
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| | - Fatima Mraiche
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, T6G 2R3 Edmonton, Canada
| | | | - Ola Hishari
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Yomna Ismail
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Jensa Joseph
- College of Pharmacy, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Maja Simonoska Crcarevska
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| | - Marija Glavas Dodov
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| | - Nikola Geskovski
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| | - Katerina Goracinova
- Institute of Pharmaceutical Technology, Faculty of Pharmacy, University of Ss. Cyril and Methodius in Skopje, 1000 Skopje, North Macedonia
| |
Collapse
|
9
|
Wang Y, Tang SC. The race to develop oral SERDs and other novel estrogen receptor inhibitors: recent clinical trial results and impact on treatment options. Cancer Metastasis Rev 2022; 41:975-990. [PMID: 36229710 PMCID: PMC9560885 DOI: 10.1007/s10555-022-10066-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/29/2022] [Indexed: 01/25/2023]
Abstract
Hormonal therapy plays a vital part in the treatment of estrogen receptor-positive (ER +) breast cancer. ER can be activated in a ligand-dependent and independent manner. Currently available ER-targeting agents include selective estrogen receptor modulators (SERMs), selective estrogen receptor degraders (SERDs), and aromatase inhibitors (AIs). Estrogen receptor mutation (ESR1 mutation) is one of the common mechanisms by which breast cancer becomes resistant to additional therapies from SERMs or AIs. These tumors remain sensitive to SERDs such as fulvestrant. Fulvestrant is limited in clinical utilization by its intramuscular formulation and once-monthly injection in large volumes. Oral SERDs are being rapidly developed to replace fulvestrant with the potential of higher efficacy and lower toxicities. Elacestrant is the first oral SERD that went through a randomized phase III trial showing increased efficacy, especially in tumors bearing ESR1 mutation, and good tolerability. Two other oral SERDs recently failed to achieve the primary endpoints of longer progression-free survival (PFS). They targeted tumors previously treated with several lines of prior therapies untested for ESR1 mutation. Initial clinical trial data demonstrated that tumors without the ESR1 mutation are less likely to benefit from the SERDs and may still respond to SERMs or AIs, including tumors previously exposed to hormonal therapy. Testing for ESR1 mutation in ongoing clinical trials and in hormonal therapy for breast cancer is highly recommended. Novel protein degradation technologies such as proteolysis-targeting chimera (PROTACS), molecular glue degrader (MGD), and lysosome-targeting chimeras (LYTACS) may result in more efficient ER degradation, while ribonuclease-targeting chimeras (RIBOTAC) and small interfering RNA (siRNA) may inhibit the production of ER protein.
Collapse
Affiliation(s)
- Yating Wang
- Ascension Providence Hospital, Southfield, MI, USA
| | - Shou-Ching Tang
- Cancer Center and Research Institute, University of Mississippi Medical Center, Guyton Research Building, G-651-07, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
10
|
Dhanya CR, Mary AS, Madhavan M. Aptamer-siRNA chimeras: Promising tools for targeting HER2 signaling in cancer. Chem Biol Drug Des 2022; 101:1162-1180. [PMID: 36099164 DOI: 10.1111/cbdd.14143] [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: 12/19/2021] [Revised: 08/28/2022] [Accepted: 09/11/2022] [Indexed: 11/30/2022]
Abstract
RNA interference is a transformative approach and has great potential in the development of novel and more efficient cancer therapeutics. Immense prospects exist in the silencing of HER2 and its downstream genes which are overexpressed in many cancers, through exogenously delivered siRNA. However, there is still a long way to exploit the full potential and versatility of siRNA therapeutics due to the challenges associated with the stability and delivery of siRNA targeted to specific sites. Aptamers offer several advantages as a vehicle for siRNA delivery, over other carriers such as antibodies. In this review, we discuss the progress made in the development and applications of aptamer-siRNA chimeras in HER2 targeting and gene silencing. A schematic workflow is also provided which will provide ample insight for all those researchers who are new to this field. Also, we think that a mechanistic understanding of the HER2 signaling pathway is crucial in designing extensive investigations aimed at the silencing of a wider array of genes. This review is expected to stimulate more research on aptamer-siRNA chimeras targeted against HER2 which might arm us with potential effective therapeutic interventions for the management of cancer.
Collapse
Affiliation(s)
- C R Dhanya
- Department of Biochemistry, Government College Kariavattom, Thiruvananthapuram, Kerala, India
| | - Aarcha Shanmugha Mary
- Department of Microbiology, Central University of Tamil Nadu, Thiruvarur, Tamil Nadu, India
| | - Maya Madhavan
- Department of Biochemistry, Government College for Women, Thiruvananthapuram, Kerala, India
| |
Collapse
|
11
|
Li X, Mei W, Wu Q, Wang J, Qi L. Theranostic Ruthenium Polypyridine Nanoparticles for Targeted Chimera Delivery into Ovarian Cancer Cells. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Efficient in vivo delivery of small interfering RNAs (siRNAs) to target cells is challenging in clinical applications. Ruthenium (II) polypyridyl complexes have been discovered as imaging theranostic and anticancer agents due to their photophysical and biological properties.
However, the clinical implementation of ruthenium complexes is limited by cancer cell selectivity. This study presents a novel siRNA delivery nanoplatform by ruthenium polypyridine complex nanoparticles (RPNs). The EGFR RNA aptamer and Notch3 siRNA chimera-loaded RPNs showed
superior RNAi effects against Notch3 gene compared to Lipofectamine. Also, RPN-chimera complexes exhibited significant in vivo antitumor effects against ovarian cancer, which exhibited much potential in future cancer imaging guided gene therapy.
Collapse
Affiliation(s)
- Xia Li
- Shanghai East Hospital, Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| | - Wenjie Mei
- Department of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Qiong Wu
- Department of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jianjun Wang
- Department of Obstetrics and Gynecology, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, 200120, China
| | - Lifeng Qi
- Shanghai East Hospital, Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, 200120, China
| |
Collapse
|
12
|
Yuhan J, Zhu L, Zhu L, Huang K, He X, Xu W. Cell-specific aptamers as potential drugs in therapeutic applications: A review of current progress. J Control Release 2022; 346:405-420. [PMID: 35489545 DOI: 10.1016/j.jconrel.2022.04.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/23/2022] [Accepted: 04/24/2022] [Indexed: 12/23/2022]
Abstract
Cell-specific aptamers are a promising emerging player in the field of disease therapy. This paper reviews the multidimensional research progress made in terms of their classification, modification, and application. Based on the target location of cell-specific aptamers, it is defined and classified cell-specific aptamers into three groups including aptamers for cell surface markers, aptamers for intracellular components, and aptamers for extracellular components. Moreover, the modification methods of aptamers to achieve improved stability and affinity are concluded. In addition, recent advances in the application of cell-specific aptamers are discussed, mainly focusing on the increasing research attraction of cell state improving helpers and cell recruitment mediators in the improvement of cellular microenvironments to achieve successful disease therapy. This review also highlights 11 types of clinical aptamer drugs. Finally, the challenges and future directions of potential clinical applications are presented. In summary, we believe that cell-specific aptamers are promising drugs in disease therapy.
Collapse
Affiliation(s)
- Jieyu Yuhan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Liye Zhu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Longjiao Zhu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Kunlun Huang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xiaoyun He
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Wentao Xu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
13
|
Wynn CS, Tang SC. Anti-HER2 therapy in metastatic breast cancer: many choices and future directions. Cancer Metastasis Rev 2022; 41:193-209. [PMID: 35142964 PMCID: PMC8924093 DOI: 10.1007/s10555-022-10021-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/19/2022] [Indexed: 12/30/2022]
Abstract
Metastatic HER2 + breast cancer is an expanding area of drug development and research, with three new drugs approved in 2020 alone. While first-line therapy is well-established for metastatic HER2 + breast cancer, the standard of care for second-line therapy will likely be changing soon based on the results of the DESTINY-Breast03 trial. In the third-line setting, many options are available. Considerations in choosing between regimens in the third-line include resistance to trastuzumab, the presence of brain metastases, and tolerability. High rates of resistance exist in this setting particularly due to expression of p95, a truncated form of HER2 that constitutively activates downstream signaling pathways. We suggest a tyrosine kinase inhibitor (TKI)-based regimen because of the activity of TKIs in brain metastases and in p95-expressing tumors. Attempts to overcome resistance to anti-HER2 therapies with PI3K inhibitors, mTOR inhibitors, and CDK 4/6 inhibitors are an active area of research. In the future, biomarkers are needed to help predict which therapies patients may benefit from the most. We review the many new drugs in development, including those with novel mechanisms of action.
Collapse
Affiliation(s)
- Carrie S Wynn
- Cancer Center and Research Institute, University of Mississippi Medical Center, Guyton Research Building, G-651-07, 2500 North State Street, Jackson, MS, 39216, USA
| | - Shou-Ching Tang
- Cancer Center and Research Institute, University of Mississippi Medical Center, Guyton Research Building, G-651-07, 2500 North State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
14
|
Qin H, Teng R, Liu Y, Li J, Yu M. Drug Release from Gelsolin-Targeted Phase-Transition Nanoparticles Triggered by Low-Intensity Focused Ultrasound. Int J Nanomedicine 2022; 17:61-71. [PMID: 35023919 PMCID: PMC8747719 DOI: 10.2147/ijn.s341421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/26/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose Current strategies for tumour-induced sentinel lymph node detection and metastasis therapy have limitations. It is essential to identify and provide warnings earlier for tumour metastasis to carry out effective clinical interventions. In addition, traditional cancer chemotherapy encounters drastic limitations due to the nonspecific delivery of antitumour drugs and severe side effects. We aimed to exploit the potential of gelsolin (GSN) monoclonal antibody as a targeting agent and perfluorohexane (PFH) as a phase-transition agent to maximize the cytotoxic effect of poly(lactic-co-glycolic acid) (PLGA) nanoparticle-based drug controllable release systems for Hca-F cells. Methods We co-encapsulated PFH and doxorubicin (DOX) into PLGA nanoparticles (NPs) and further conjugated GSN monoclonal antibody onto the surface of NPs to form GSN-targeted phase transition polymer NPs (GSN-PLGA-PFH-DOX) for both imaging and therapy of tumours and metastatic lymph nodes. To promote and trigger drug release on demand, low-intensity focused ultrasound (LIFU) was applied to achieve a controllable release of the encapsulated drug. Results GSN-PLGA-PFH-DOX NPs exhibited characteristics such as a narrow size distribution and smooth surface. GSN-PLGA-PFH-DOX NPs could also specifically bind to Hca-F cells and increase the ultrasound contrast agent (UCA) image contrast intensity. GSN-PLGA-PFH-DOX NPs enable GSN-mediated targeting and biotherapeutic effects as well as LIFU-responsive drug release, resulting in synergistic cytotoxic effects in GSN-overexpressing cells in vitro. Conclusion Our work might provide a strategy for the imaging and chemotherapy of primary tumours and their metastases.
Collapse
Affiliation(s)
- Haocheng Qin
- Department of Ultrasound, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, People's Republic of China
| | - Rong Teng
- Department of Ultrasound, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, People's Republic of China
| | - Yan Liu
- Department of Ultrasound, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, People's Republic of China
| | - Juan Li
- Department of Oncology, The Second People's Hospital of Lianyungang, Lianyungang, 222023, People's Republic of China
| | - Ming Yu
- Department of Ultrasound, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222002, People's Republic of China
| |
Collapse
|
15
|
Mansour MA, Caputo VS, Aleem E. Highlights on selected growth factors and their receptors as promising anticancer drug targets. Int J Biochem Cell Biol 2021; 140:106087. [PMID: 34563698 DOI: 10.1016/j.biocel.2021.106087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
Growth factor receptors (GFRs) and receptor tyrosine kinases (RTK) are groups of proteins mediating a plethora of physiological processes, including cell growth, proliferation, survival, differentiation and migration. Under certain circumstances, expression of GFRs and subsequently their downstream kinase signaling are deregulated by genetic, epigenetic, and somatic changes leading to uncontrolled cell division in many human diseases, most notably cancer. Cancer cells rely on growth factors to sustain the increasing need to cell division and metabolic reprogramming through cancer-associated activating mutations of their receptors (i.e., GFRs). In this review, we highlight the recent advances of selected GFRs and their ligands (growth factors) in cancer with emphasis on structural and functional differences. We also interrogate how overexpression and/or hyperactivation of GFRs contribute to cancer initiation, development, progression, and resistance to conventional chemo- and radiotherapies. Novel approaches are being developed as anticancer agents to target growth factor receptors and their signaling pathways in different cancers. Here, we illustrate how the current knowledge of GFRs biology, and their ligands lead to development of targeted therapies to inhibit and/or block the activity of growth factors, GFRs and downstream kinases to treat diseases such as cancer.
Collapse
Affiliation(s)
- Mohammed A Mansour
- Cancer Biology and Therapy Lab, Division of Human Sciences, School of Applied Sciences, London South Bank University, London, UK; Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Valentina S Caputo
- Cancer Biology and Therapy Lab, Division of Human Sciences, School of Applied Sciences, London South Bank University, London, UK
| | - Eiman Aleem
- Cancer Biology and Therapy Lab, Division of Human Sciences, School of Applied Sciences, London South Bank University, London, UK.
| |
Collapse
|
16
|
He Q, Liu Y, Li K, Wu Y, Wang T, Tan Y, Jiang T, Liu X, Liu Z. Deoxyribonucleic acid anchored on cell membranes for biomedical application. Biomater Sci 2021; 9:6691-6717. [PMID: 34494042 DOI: 10.1039/d1bm01057c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Engineering cellular membranes with functional molecules provides an attractive strategy to manipulate cellular behaviors and functionalities. Currently, synthetic deoxyribonucleic acid (DNA) has emerged as a promising molecular tool to engineer cellular membranes for biomedical applications due to its molecular recognition and programmable properties. In this review, we summarized the recent advances in anchoring DNA on the cellular membranes and their applications. The strategies for anchoring DNA on cell membranes were summarized. Then their applications, such as immune response activation, receptor oligomerization regulation, membrane structure mimicking, cell-surface biosensing, and construction of cell clusters, were listed. The DNA-enabled intelligent systems which were able to sense stimuli such as DNA strands, light, and metal ions were highlighted. Finally, insights regarding the remaining challenges and possible future directions were provided.
Collapse
Affiliation(s)
- Qunye He
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China.
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan Province, P. R. China
| | - Ke Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China.
| | - Yuwei Wu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China.
| | - Ting Wang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China.
| | - Yifu Tan
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan Province, P. R. China
| | - Ting Jiang
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan Province, P. R. China
| | - Xiaoqin Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, Hunan Province, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan Province, P. R. China. .,Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan, P. R. China
| |
Collapse
|
17
|
Vi C, Mandarano G, Shigdar S. Diagnostics and Therapeutics in Targeting HER2 Breast Cancer: A Novel Approach. Int J Mol Sci 2021; 22:6163. [PMID: 34200484 PMCID: PMC8201268 DOI: 10.3390/ijms22116163] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/25/2021] [Accepted: 05/30/2021] [Indexed: 01/02/2023] Open
Abstract
Breast cancer is one of the most commonly occurring cancers in women globally and is the primary cause of cancer mortality in females. BC is highly heterogeneous with various phenotypic expressions. The overexpression of HER2 is responsible for 15-30% of all invasive BC and is strongly associated with malignant behaviours, poor prognosis and decline in overall survival. Molecular imaging offers advantages over conventional imaging modalities, as it provides more sensitive and specific detection of tumours, as these techniques measure the biological and physiological processes at the cellular level to visualise the disease. Early detection and diagnosis of BC is crucial to improving clinical outcomes and prognosis. While HER2-specific antibodies and nanobodies may improve the sensitivity and specificity of molecular imaging, the radioisotope conjugation process may interfere with and may compromise their binding functionalities. Aptamers are single-stranded oligonucleotides capable of targeting biomarkers with remarkable binding specificity and affinity. Aptamers can be functionalised with radioisotopes without compromising target specificity. The attachment of different radioisotopes can determine the aptamer's functionality in the treatment of HER2(+) BC. Several HER2 aptamers and investigations of them have been described and evaluated in this paper. We also provide recommendations for future studies with HER2 aptamers to target HER2(+) BC.
Collapse
Affiliation(s)
- Chris Vi
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (C.V.); (G.M.)
| | - Giovanni Mandarano
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (C.V.); (G.M.)
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| | - Sarah Shigdar
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia; (C.V.); (G.M.)
- Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| |
Collapse
|
18
|
Jin H. Perspectives of Aptamers for Medical Applications. APTAMERS FOR MEDICAL APPLICATIONS 2021:405-462. [DOI: 10.1007/978-981-33-4838-7_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
19
|
Jain S, Kaur J, Prasad S, Roy I. Nucleic acid therapeutics: a focus on the development of aptamers. Expert Opin Drug Discov 2020; 16:255-274. [PMID: 32990095 DOI: 10.1080/17460441.2021.1829587] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Aptamers provide exciting opportunities for the development of specific and targeted therapeutic approaches. AREAS COVERED In this review, the authors discuss different therapeutic options available with nucleic acids, including aptamers, focussing on similarities and differences between them. The authors concentrate on case studies with specific aptamers, which exemplify their distinct advantages. The reasons for failure, wherever available, are deliberated upon. Attempts to accelerate the in vitro selection process have been discussed. Challenges with aptamers in terms of their specificity and targeted delivery and strategies to overcome these are described. Examples of precise regulation of systemic half-life of aptamers using antidotes are discussed. EXPERT OPINION Despite their nontoxic nature, a variety of reasons limit the therapeutic potential of aptamers in the clinic. The analysis of adverse effects observed with the pegnivacogin/anivamersen pair has highlighted the need to screen for preexisting PEG antibodies in any clinical trial involving pegylated molecules. Surprisingly, and promisingly, the ability of nucleic acid therapeutics to breach the blood brain barrier seems achievable. The recognition of specific motifs, e.g. G-quadruplex in thrombin-binding aptamers, or a 'nucleation' zone while designing aptamer-antidote pairs, is likely to accelerate the discovery of therapeutically efficacious molecules.
Collapse
Affiliation(s)
- Swati Jain
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | - Jaskirat Kaur
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | - Shivcharan Prasad
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Punjab, India
| | - Ipsita Roy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Punjab, India
| |
Collapse
|
20
|
Ma GL, Qiao ZL, He D, Wang J, Kong YY, Xin XY, Wen FQ, Bao SJ, Ma ZR, Wang FS, Xie J, Hu YH. Establishment of a low-tumorigenic MDCK cell line and study of differential molecular networks. Biologicals 2020; 68:112-121. [PMID: 32928630 DOI: 10.1016/j.biologicals.2020.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/11/2020] [Accepted: 07/14/2020] [Indexed: 12/18/2022] Open
Abstract
Influenza is an acute respiratory infection caused by the influenza virus, and vaccination against influenza is considered the best way to prevent the onset and spread. MDCK (Madin-Darby canine kidney) cells are typically used to isolate the influenza virus, however, their high tumorigenicity is the main controversy in the production of influenza vaccines. Here, MDCK-C09 and MDCK-C35 monoclonal cell lines were established, which were proven to be low in tumorigenicity. RNA-seq of MDCK-C09, MDCK-C35, and MDCK-W73 cells was performed to investigate the putative tumorigenicity mechanisms. Tumor-related molecular interaction analysis of the differentially expressed genes indicates that hub genes, such as CUL3 and EGFR, may play essential roles in tumorigenicity differences between MDCK-C (MDCK-C09 and MDCK-C35) and MDCK-W (MDCK-W73) cells. Moreover, the analysis of cell proliferation regulation-associated molecular interaction shows that downregulated JUN and MYC, for instance, mediate increased proliferation of these cells. The present study provides a new low-tumorigenic MDCK cell line and describes the potential molecular mechanism for the low tumorigenicity and high proliferation rate.
Collapse
Affiliation(s)
- Gui-Lan Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730030, PR China; Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, PR China.
| | - Zi-Lin Qiao
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, PR China.
| | - Dan He
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, PR China.
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China.
| | - Yan-Yan Kong
- Huashan Hospital Affiliated to Fudan University, Shanghai, China.
| | - Xiao-Yong Xin
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730030, PR China.
| | - Feng-Qin Wen
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730030, PR China.
| | - Shi-Jun Bao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730030, PR China.
| | - Zhong-Ren Ma
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, PR China.
| | - Fu-Shuai Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China.
| | - Jiang Xie
- School of Computer Engineering and Science, Shanghai University, Shanghai, China.
| | - Yong-Hao Hu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, 730030, PR China.
| |
Collapse
|
21
|
Bhatia P, Sharma V, Alam O, Manaithiya A, Alam P, Kahksha, Alam MT, Imran M. Novel quinazoline-based EGFR kinase inhibitors: A review focussing on SAR and molecular docking studies (2015-2019). Eur J Med Chem 2020; 204:112640. [PMID: 32739648 DOI: 10.1016/j.ejmech.2020.112640] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022]
Abstract
The over expression of EGFR has been recognized as the driver mechanism in the occurrence and progression of carcinomas such as lung cancer, breast cancer, pancreatic cancer, etcetera. EGFR receptor was thus established as an important target for the management of solid tumors. The occurrence of resistance caused as a result of mutations in EGFR has presented a formidable challenge in the discovery of novel inhibitors of EGFR. This has resulted in the development of three generations of EGFR TKIs. Newer mutations like C797S cause failure of Osimertinib and other EGFR TKIs belonging to the third-generation caused by the development of resistance. In this review, we have summarized the work done in the last five years to overcome the limitations of currently marketed drugs, giving structural activity relationships of quinazoline-based lead compounds synthesized and tested recently. We have also highlighted the shortcomings of the currently used approaches and have provided guidance for circumventing these limitations. Our review would help medicinal chemists streamline and guide their efforts towards developing novel quinazoline-based EGFR inhibitors.
Collapse
Affiliation(s)
- Parth Bhatia
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Vrinda Sharma
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Ozair Alam
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Ajay Manaithiya
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Perwaiz Alam
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Kahksha
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Md Tauquir Alam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha, Pin Code 91911, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha, Pin Code 91911, Saudi Arabia
| |
Collapse
|
22
|
Zhang L, Mu C, Zhang T, Wang Y, Wang Y, Fan L, Liu C, Chen H, Shen J, Wei K, Li H. Systemic Delivery of Aptamer-Conjugated XBP1 siRNA Nanoparticles for Efficient Suppression of HER2+ Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2020; 12:32360-32371. [PMID: 32613835 DOI: 10.1021/acsami.0c07353] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
siRNA therapeutics as an emerging class of drug development is successfully coming to clinical utilization. The RNA-based therapy is widely utilized to explore the mechanism and cure a variety of gene-specific diseases. Tumor is an oncogene-driven disease; many genes are related to tumor progression and chemoresistance. Although human epidermal growth factor receptor 2 (HER2)-targeted monoclonal antibody therapy has dramatically improved the survival rate, chemotherapy remains essential to HER2-positive (HER2+) breast cancer patients. Recently, X-box binding protein 1 (XBP1) has been involved in triple-negative breast cancer (TNBC) chemoresistance and progression, but its function in HER2+ breast cancer is poorly explored. Here, we silenced XBP1 expression using RNase-resistant RNA nanoparticles (NPs). Intravenous injection of RNA NPs with HER2-specific aptamers resulted in strong binding to tumors but not to healthy tissues. XBP1 deletion by RNA NPs impaired angiogenesis and inhibited cell proliferation, significantly suppressed breast cancer growth, and promoted the sensitization of chemotherapy in an HER2+ breast cancer mouse model. Overall, these results reveal the function of XBP1 in HER2+ breast cancer development and chemoresistance and imply that targeting XBP1 by RNA NPs may offer an easy and promising strategy for a combination treatment of breast cancer in the future.
Collapse
Affiliation(s)
- Long Zhang
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, P. R. China
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province 325011, P. R. China
| | - Chaofeng Mu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, P. R. China
| | - Tinghong Zhang
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, P. R. China
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province 325011, P. R. China
| | - Yingying Wang
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, P. R. China
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province 325011, P. R. China
| | - Yili Wang
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, P. R. China
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province 325011, P. R. China
| | - Luhui Fan
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, P. R. China
| | - Cong Liu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, P. R. China
| | - Hao Chen
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, P. R. China
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province 325011, P. R. China
| | - Jianliang Shen
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, P. R. China
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province 325011, P. R. China
| | - Kun Wei
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, P. R. China
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province 325011, P. R. China
| | - Huaqiong Li
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, P. R. China
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang Province 325011, P. R. China
| |
Collapse
|
23
|
Fu Z, Xiang J. Aptamers, the Nucleic Acid Antibodies, in Cancer Therapy. Int J Mol Sci 2020; 21:ijms21082793. [PMID: 32316469 PMCID: PMC7215806 DOI: 10.3390/ijms21082793] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023] Open
Abstract
The arrival of the monoclonal antibody (mAb) technology in the 1970s brought with it the hope of conquering cancers to the medical community. However, mAbs, on the whole, did not achieve the expected wonder in cancer therapy although they do have demonstrated successfulness in the treatment of a few types of cancers. In 1990, another technology of making biomolecules capable of specific binding appeared. This technique, systematic evolution of ligands by exponential enrichment (SELEX), can make aptamers, single-stranded DNAs or RNAs that bind targets with high specificity and affinity. Aptamers have some advantages over mAbs in therapeutic uses particularly because they have little or no immunogenicity, which means the feasibility of repeated use and fewer side effects. In this review, the general properties of the aptamer, the advantages and limitations of aptamers, the principle and procedure of aptamer production with SELEX, particularly the undergoing studies in aptamers for cancer therapy, and selected anticancer aptamers that have entered clinical trials or are under active investigations are summarized.
Collapse
Affiliation(s)
- Zhaoying Fu
- Department of Biochemistry and Molecular Biology, College of Medicine, Yanan University, Yanan 716000, China
- Correspondence: (Z.F.); (J.X.)
| | - Jim Xiang
- Division of Oncology, University of Saskatchewan, Saskatoon, SA S7N 4H4, Canada
- Correspondence: (Z.F.); (J.X.)
| |
Collapse
|
24
|
Yeldell SB, Seitz O. Nucleic acid constructs for the interrogation of multivalent protein interactions. Chem Soc Rev 2020; 49:6848-6865. [DOI: 10.1039/d0cs00518e] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Sequence-programmed self-assembly provides multivalent nucleic acid–ligand constructs used as tailor-made probes for unravelling and exploiting the mechanisms of multivalency-enhanced interactions on protein receptors.
Collapse
Affiliation(s)
- Sean B. Yeldell
- Department of Chemistry
- Humboldt-Universität zu Berlin
- Brook-Taylor-Str. 2
- 12489 Berlin
- Germany
| | - Oliver Seitz
- Department of Chemistry
- Humboldt-Universität zu Berlin
- Brook-Taylor-Str. 2
- 12489 Berlin
- Germany
| |
Collapse
|
25
|
Dinis Ano Bom AP, da Costa Neves PC, Bonacossa de Almeida CE, Silva D, Missailidis S. Aptamers as Delivery Agents of siRNA and Chimeric Formulations for the Treatment of Cancer. Pharmaceutics 2019; 11:pharmaceutics11120684. [PMID: 31888119 PMCID: PMC6956146 DOI: 10.3390/pharmaceutics11120684] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/24/2019] [Accepted: 10/30/2019] [Indexed: 12/31/2022] Open
Abstract
Both aptamers and siRNA technologies have now reached maturity, and both have been validated with a product in the market. However, although pegaptanib reached the market some time ago, there has been a slow process for new aptamers to follow. Today, some 40 aptamers are in the market, but many in combination with siRNAs, in the form of specific delivery agents. This combination offers the potential to explore the high affinity and specificity of aptamers, the silencing power of siRNA, and, at times, the cytotoxicity of chemotherapy molecules in powerful combinations that promise to delivery new and potent therapies. In this review, we report new developments in the field, following up from our previous work, more specifically on the use of aptamers as delivery agents of siRNA in nanoparticle formulations, alone or in combination with chemotherapy, for the treatment of cancer.
Collapse
Affiliation(s)
- Ana Paula Dinis Ano Bom
- Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz. Av. Brasil, 4365-Manguinhos, Rio de Janeiro/RJ CEP 21040-900, Brazil; (A.P.D.A.B.); (P.C.d.C.N.); (D.S.)
| | - Patrícia Cristina da Costa Neves
- Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz. Av. Brasil, 4365-Manguinhos, Rio de Janeiro/RJ CEP 21040-900, Brazil; (A.P.D.A.B.); (P.C.d.C.N.); (D.S.)
| | - Carlos Eduardo Bonacossa de Almeida
- Laboratório de Radiobiologia, Divisão de Física Médica, Instituto de Radioproteção e Dosimetria, Comissão Nacional de Energia Nuclear. Av. Salvador Allende S/N., Rio de Janeiro/RJ CEP 22783-127, Brazil;
| | - Dilson Silva
- Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz. Av. Brasil, 4365-Manguinhos, Rio de Janeiro/RJ CEP 21040-900, Brazil; (A.P.D.A.B.); (P.C.d.C.N.); (D.S.)
| | - Sotiris Missailidis
- Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz. Av. Brasil, 4365-Manguinhos, Rio de Janeiro/RJ CEP 21040-900, Brazil; (A.P.D.A.B.); (P.C.d.C.N.); (D.S.)
- Correspondence:
| |
Collapse
|
26
|
Characterization of A Bifunctional Synthetic RNA Aptamer and A Truncated Form for Ability to Inhibit Growth of Non-Small Cell Lung Cancer. Sci Rep 2019; 9:18836. [PMID: 31827170 PMCID: PMC6906461 DOI: 10.1038/s41598-019-55280-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/25/2019] [Indexed: 12/24/2022] Open
Abstract
An in vitro-transcribed RNA aptamer (trans-RA16) that targets non-small cell lung cancer (NSCLC) was previously identified through in vivo SELEX. Trans-RA16 can specifically target and inhibit human NCI-H460 cells in vitro and xenograft tumors in vivo. Here, in a follow-up study, we obtained a chemically-synthesized version of this RNA aptamer (syn-RA16) and a truncated form, and compared them to trans-RA16 for abilities to target and inhibit NCI-H460 cells. The syn-RA16, preferred for drug development, was by design to differ from trans-RA16 in the extents of RNA modifications by biotin, which may affect RA16’s anti-tumor effects. We observed aptamer binding to NCI-H460 cells with KD values of 24.75 ± 2.28 nM and 12.14 ± 1.46 nM for syn-RA16 and trans-RA16, respectively. Similar to trans-RA16, syn-RA16 was capable of inhibiting NCI-H460 cell viability in a dose-dependent manner. IC50 values were 118.4 nM (n = 4) for syn-RA16 and 105.7 nM (n = 4) for trans-RA16. Further studies using syn-RA16 demonstrated its internalization into NCI-H460 cells and inhibition of NCI-H460 cell growth. Moreover, in vivo imaging demonstrated the gradual accumulation of both syn-RA16 and trans-RA16 at the grafted tumor site, and qRT-PCR showed high retention of syn-RA16 in tumor tissues. In addition, a truncated fragment of trans-RA16 (S3) was identified, which exhibited binding affinity for NCI-H460 cells with a KD value of 63.20 ± 0.91 nM and inhibited NCI-H460 cell growth by 39.32 ± 3.25% at 150 nM. These features of the syn-RA16 and S3 aptamers should facilitate the development of a novel diagnostic or treatment approach for NSCLC in clinical settings.
Collapse
|
27
|
Jin P, Liu J, Zhou Q, Li S, Liu W, Xi S. Long-term treatment with arsenite activates HER1 and HER2 through upregulating EGF, TGFα, and HSP90 in a human uroepithelial cell line. Cell Biol Toxicol 2019; 36:279-284. [PMID: 31773452 DOI: 10.1007/s10565-019-09500-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/24/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Peiyu Jin
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, 110122, Shenyang, People's Republic of China
| | - Jieyu Liu
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, 110122, Shenyang, People's Republic of China
| | - Qing Zhou
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, 110122, Shenyang, People's Republic of China
| | - Sihao Li
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, 110122, Shenyang, People's Republic of China
| | - Weijue Liu
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, 110122, Shenyang, People's Republic of China
| | - Shuhua Xi
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, 110122, Shenyang, People's Republic of China.
| |
Collapse
|
28
|
Shen Y, Li M, Liu T, Liu J, Xie Y, Zhang J, Xu S, Liu H. A dual-functional HER2 aptamer-conjugated, pH-activated mesoporous silica nanocarrier-based drug delivery system provides in vitro synergistic cytotoxicity in HER2-positive breast cancer cells. Int J Nanomedicine 2019; 14:4029-4044. [PMID: 31213813 PMCID: PMC6549788 DOI: 10.2147/ijn.s201688] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/20/2019] [Indexed: 12/19/2022] Open
Abstract
Purpose: As well as functioning as a ligand that is selectively internalized by cells overexpressing human epidermal growth factor receptor-2 (HER2), HApt can exert cytotoxic effects by inducing cross-linking and subsequent translocation of HER2 to cytoplasmic vesicles, such downregulation of HER2 inhibits cell proliferation and induces apoptosis. We aimed to exploit the potential of HApt as both a targeting agent and antagonist to maximize the efficacy of mesoporous silica nanoparticle (MSN)-based drug release systems for HER2-positive breast cancer. Materials and methods: We fabricated novel HApt aptamer-functionalized pH-sensitive β-cyclodextrin (β-CD)-capped doxorubicin (DOX)-loaded mesoporous silica nanoparticles (termed MSN-BM/CD-HApt@DOX) for targeted delivery and selective targeting of HER2-positive cells. MSN-functionalized benzimidazole (MSN-BM) was used to load and achieve pH stimuli-responsive release of the chemotherapeutic agent doxorubicin (DOX). β-cyclodextrin was introduced as a gatekeeper for encapsulated DOX and HApt as a selective HER2-targeting moiety and biotherapeutic agent. Results: Physical and chemical characterizations (FT-IR, XRD, TEM and BET) confirmed successful construction of MSN-BM/CD-HApt@DOX nanoparticles. In vitro release assays verified pH-sensitive DOX release. MSN-BM/CD-HApt@DOX (relative DOX concentration, 3.6 μg/mL) underwent HER2-mediated endocytosis and was more cytotoxic to HER2-positive SKBR3 cells than HER2-negative MCF7 cells. MSN-BM/CD-HApt@DOX also exhibited better uptake and stronger growth inhibition in SKBR3 cells than the control MSN-BM/CD-NCApt@DOX functionalized with a scrambled nucleotide sequence on CD. Overall, intracellular delivery of DOX and the biotherapeutic agent HApt resulted in synergistic cytotoxic effects in HER2-positive cancer cells in comparison to either DOX or HApt alone. Conclusion: MSN-BM/CD-HApt@DOX enables HER2-mediated targeting and biotherapeutic effects as well as pH-responsive DOX drug release, resulting in synergistic cytotoxic effects in HER2-overexpressing cells in vitro. This novel nanocarrier could potentially enable specific targeting to improve the efficacy of chemotherapy for HER2-positive cancer.
Collapse
Affiliation(s)
- Yinxing Shen
- Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, People’s Republic of China
| | - Mengya Li
- Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Tianqi Liu
- Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Jing Liu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, People’s Republic of China
| | - Youhua Xie
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, People’s Republic of China
| | - Junqi Zhang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200032, People’s Republic of China
| | - Shouhong Xu
- Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Honglai Liu
- Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| |
Collapse
|
29
|
Aptamer Chimeras for Therapeutic Delivery: The Challenging Perspectives. Genes (Basel) 2018; 9:genes9110529. [PMID: 30384431 PMCID: PMC6266988 DOI: 10.3390/genes9110529] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 12/29/2022] Open
Abstract
Nucleic acid-based aptamers have emerged as efficient delivery carriers of therapeutics. Thanks to their unique features, they can be, to date, considered one of the best targeting moieties, allowing the specific recognition of diseased cells and avoiding unwanted off-target effects on healthy tissues. In this review, we revise the most recent contributes on bispecific and multifunctional aptamer therapeutic chimeras. We will discuss key examples of aptamer-mediated delivery of nucleic acid and peptide-based therapeutics underlying their great potentiality and versatility. Achieved objectives and challenges will be highlighted as well.
Collapse
|