1
|
Jiang Y, Cao Y, Yao Y, Zhang D, Wang Y. Chitosan and hyaluronic acid in breast cancer treatment: Anticancer efficacy and nanoparticle and hydrogel development. Int J Biol Macromol 2025; 301:140144. [PMID: 39848359 DOI: 10.1016/j.ijbiomac.2025.140144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
The pervasive global health concern of breast cancer necessitates the development of innovative therapeutic interventions to enhance efficacy and mitigate adverse effects. Chitosan and hyaluronic acid, recognized for their biocompatibility and biodegradability, present compelling options for the novel drug delivery systems and therapeutic platforms in the context of breast cancer management. This review will delineate the distinctive attributes of chitosan and hyaluronic acid, encompassing their inherent anticancer properties, targeting capabilities, and suitability for chemical modifications along with nanoparticle development. These characteristics render them exceptionally well-suited for the fabrication of nanoparticles and hydrogels. The intrinsic anticancer potential of chitosan, in conjunction with its mucoadhesive properties, and the robust binding affinity of hyaluronic acid to CD44 receptors, facilitate specific drug delivery to the malignant cells, thus circumventing the limitations inherent in traditional treatment modalities such as chemotherapy. The incorporation of these materials into nanocarriers allows for the co-delivery of therapeutic agents, thereby potentiating synergistic effects, while hydrogel systems provide localized, controlled drug release and facilitate tissue regeneration. An analysis of advancements in their synthesis, functionalization, and application is presented, while also acknowledging challenges pertaining to scalability and clinical translation.
Collapse
Affiliation(s)
- Yanlin Jiang
- Department of Breast and Thyroid Surgery, the Affiliated Zhongshan Hospital of Dalian University, China
| | - Yu Cao
- Department of Surgical Oncology and Breast Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yiqun Yao
- Department of Breast and Thyroid Surgery, the Affiliated Zhongshan Hospital of Dalian University, China
| | - Dianlong Zhang
- Department of Breast and Thyroid Surgery, the Affiliated Zhongshan Hospital of Dalian University, China.
| | - Yuying Wang
- Department of Breast Surgery, The Cancer Hospital of China Medical University Liaoning Cancer Hospital & Institute, China.
| |
Collapse
|
2
|
Zhang L, Zhang H. Recent advances of affibody molecules in biomedical applications. Bioorg Med Chem 2024; 113:117923. [PMID: 39278106 DOI: 10.1016/j.bmc.2024.117923] [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: 05/20/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
Affibody molecules are 58-amino-acid peptides with a molecular weight of about 6.5 kDa, derived from the Z domain of Staphylococcal Protein A. Since they have been used as substitutes for antibodies in biomedicine, several therapeutic affibody molecules have been developed for clinical use. Additionally, affibody molecules have been designed for a range of different applications. This review focuses on the progress made in the last five years in the field of affibody molecules and their potential uses in medical imaging, especially in oncology and cancer treatment. It covers areas such as molecular imaging, targeted delivery of toxic drugs, and their use in combination with nanoparticles. We also highlight some current biomedical applications where affibody molecules are commonly used as a "guide." Due to their many advantages, affibody molecules offer significant potential for applications in both biochemical and medical fields.
Collapse
Affiliation(s)
- Liuyanlin Zhang
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, MOE Key Laboratory of Molecular Biophysics, Wuhan 430074, China
| | - Houjin Zhang
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, MOE Key Laboratory of Molecular Biophysics, Wuhan 430074, China.
| |
Collapse
|
3
|
Park JY, Kang M, Lim S, Cho H, Yang S, Baek SY, Tan L, Song C, Lee M, Yeom B, Ha JS, Lee S, Kim Y. Assembly of 2′,3′-Cyclic guanosine Monophosphate-Adenosine monophosphate and their spontaneous intracellular disassembly for enhanced antitumor immunity via natural STING pathway activation. CHEMICAL ENGINEERING JOURNAL 2024; 500:157037. [DOI: 10.1016/j.cej.2024.157037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Li N, Chen S, Cai X. Harnessing molecular probes for imaging of human epidermal growth factor receptor (HER) family. Bioorg Med Chem 2024; 113:117931. [PMID: 39362074 DOI: 10.1016/j.bmc.2024.117931] [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: 08/18/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024]
Abstract
The human epidermal growth factor receptor (HER) family plays a critical role in the development, migration, and invasion of various cancers. Currently, the FDA has approved numerous targeting therapies for the HER family consist of small molecule drugs, monoclonal antibodies and antibody-drug conjugates. To facilitate precision therapy using currently approved targeted agents, early detection and quantification of each HER receptor are essential for assessment, treatment, and prognostic purposes. This study provides a comprehensive review of the latest advancements in detection and quantification of HER receptors, including traditional biopsies, liquid biopsies, and non-invasive detection methods. Although traditional histological methods, such as immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH), have yielded valuable insights, advancements in real-time and non-invasive detection technologies necessitate improved methods for the dynamic evaluation of HER status. This article also reviews several emerging real-time techniques for detecting and quantifying HER status in circulating tumor cells (CTCs) extracted from blood samples, as well as in vivo assessments using positron emission tomography (PET) and single-photon emission computed tomography (SPECT) imaging. This review emphasizes the importance of continuous innovation in the application of HER receptor imaging technologies, with the goal of enhancing treatment outcomes and prognoses for cancer patients.
Collapse
Affiliation(s)
- Na Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China
| | - Shengxi Chen
- Biodesign Center for BioEnergetics, Arizona State University, Tempe 85287, USA.
| | - Xiaoqing Cai
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China.
| |
Collapse
|
5
|
Kumar M, Jha A, Mishra B. DNA-Based Nanostructured Platforms as Drug Delivery Systems. CHEM & BIO ENGINEERING 2024; 1:179-198. [PMID: 39974200 PMCID: PMC11835166 DOI: 10.1021/cbe.3c00023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/17/2023] [Accepted: 12/17/2023] [Indexed: 02/21/2025]
Abstract
DNA nanotechnology has recently provided a novel approach for developing safe, biocompatible, biodegradable, non-immunogenic, and non-toxic drug delivery systems. DNA nanostructures have numerous advantages for deployment as drug delivery platforms, owing to programmable assembly, ease of production, reproducibility, and precise control over size, shape, and function. DNA nanostructures can dramatically improve the delivery of poorly soluble drugs, decreasing cytotoxicity to normal tissues and improving therapeutic effectiveness. Using different conjugation methods, DNA nanostructures can be precisely integrated with a wide range of functional moieties, including proteins, peptides, aptamers, polymers, lipids, inorganic nanoparticles, and targeting groups, to enhance the nanostructure stability, extend circulation, and specify drug delivery. Smart DNA nanostructures with targeting ligands or a stimuli-responsive moiety specify therapeutic delivery to target, minimize drug loss attributable to prior drug release or off-target distribution, improve target accumulation, promote cellular internalization, bypass efflux pumps, and avoid adverse effects. Target-specific delivery by smart DNA nanostructures, in turn, maximizes drug concentration, reaching the target locations at a faster rate and preventing therapeutic failure while also lowering the dose needed for therapeutic effect. This Review provides an overview of DNA nanostructures for drug encapsulation and selective delivery to the desired sites. DNA nanostructures are a novel platform for drug delivery with improved performance that may be used to treat a variety of diseases.
Collapse
Affiliation(s)
| | | | - Brahmeshwar Mishra
- Department of Pharmaceutical Engineering
& Technology, Indian Institute of Technology,
(BHU), Varanasi 221005, Uttar Pradesh, India
| |
Collapse
|
6
|
Yu L, Xu Y, Al-Amin M, Jiang S, Sample M, Prasad A, Stephanopoulos N, Šulc P, Yan H. CytoDirect: A Nucleic Acid Nanodevice for Specific and Efficient Delivery of Functional Payloads to the Cytoplasm. J Am Chem Soc 2023; 145:27336-27347. [PMID: 38055928 PMCID: PMC10789493 DOI: 10.1021/jacs.3c07491] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Direct and efficient delivery of functional payloads such as chemotherapy drugs, siRNA, or small-molecule inhibitors into the cytoplasm, bypassing the endo/lysosomal trapping, is a challenging task for intracellular medicine. Here, we take advantage of the programmability of DNA nanotechnology to develop a DNA nanodevice called CytoDirect, which incorporates disulfide units and human epidermal growth factor receptor 2 (HER2) affibodies into a DNA origami nanostructure, enabling rapid cytosolic uptake into targeted cancer cells and deep tissue penetration. We further demonstrated that therapeutic oligonucleotides and small-molecule chemotherapy drugs can be easily delivered by CytoDirect and showed notable effects on gene knockdown and cell apoptosis, respectively. This study demonstrates the synergistic effect of disulfide and HER2 affibody modifications on the rapid cytosolic delivery of DNA origami and its payloads to targeted cells and deep tissues, thereby expanding the delivery capabilities of DNA nanostructures in a new direction for disease treatment.
Collapse
Affiliation(s)
- Lu Yu
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Yang Xu
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Md Al-Amin
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Shuoxing Jiang
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Matthew Sample
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Abhay Prasad
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Nicholas Stephanopoulos
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Petr Šulc
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| | - Hao Yan
- School of Molecular Sciences and Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, Arizona 85281, United States
| |
Collapse
|
7
|
Cai X, Zhang L, Chen S. Editorial: Cancer treatment and early detection targeting HER receptors, Volume II. Front Mol Biosci 2023; 10:1229765. [PMID: 37426424 PMCID: PMC10325823 DOI: 10.3389/fmolb.2023.1229765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023] Open
Affiliation(s)
- Xiaoqing Cai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Libing Zhang
- Tianjin Key Laboratory of Molecular Optoelectronic, Department of Chemistry, Tianjin University, Tianjin, China
| | - Shengxi Chen
- Biodesign Center for Bioenergetics, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
8
|
Peng Y, Wu Z, Pang Z, Zhang L, Song D, Liu F, Li Y, Lin T. Manufacture and evaluation of a HER2-positive breast cancer immunotoxin 4D5Fv-PE25. Microb Cell Fact 2023; 22:100. [PMID: 37198642 DOI: 10.1186/s12934-023-02115-0] [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: 10/08/2022] [Accepted: 05/10/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND Human epidermal growth factor receptor 2 (HER2) positive breast cancer is an aggressive subtype, accounting for around 20% of all breast cancers. The development of HER2-targeted therapy has substantially improved patient outcomes. Nevertheless, the increasing rate of side effects and resistance to targeted drugs limit their efficacy in clinical practice. In this study, we designed and synthesized a new immunotoxin, 4D5Fv-PE25, which targets HER2-positive breast cancer, and evaluated its effectiveness in vitro and in vivo. RESULTS The 4D5Fv-PE25 was expressed in high-density Escherichia coli (E. coli.) using the fermentor method and refined via hydrophobicity, ion exchange, and filtration chromatography, achieving a 56.06% recovery rate. Additionally, the semi-manufactured product with 96% purity was prepared into freeze-dried powder by the lyophilized process. Flow cytometry was used to detect the expression of HER2 in SK-BR-3, BT-474, MDA-MB-231, and MDA-MB-468 breast cancer cell lines. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) method was used for cytotoxicity assay, and the half-maximal inhibitory concentration (IC50) of 4D5Fv-PE25 lyophilized products to HER2-positive cell line SK-BR-3 was 12.53 ng/mL. The 4D5Fv-PE25 was injected into xenograft tumor mice via the tail vein on the 1st, 4th, and 8th day, it indicated that the growth of tumor volume was effectively inhibited for 24 days, although the 4D5Fv-PE25 was metabolized within 60 min by measuring the release of 3 H-Thymidine radiation. CONCLUSION we succeeded in producing the 4D5Fv-PE25 freeze-dried powder using the prokaryotic expression method, and it could be employed as a potential drug for treating HER2-positive breast cancer.
Collapse
Affiliation(s)
- Yanjie Peng
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
| | - Zhengli Wu
- Panacea Bioscience inc, Halifax, NS, Canada
- College of Fisheries, Southwest University, Beibei, Chongqing, 400715, China
| | - Zheng Pang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, 250300, China
| | - Lin Zhang
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Dandan Song
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Fang Liu
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Yanhong Li
- Panacea Bioscience inc, Halifax, NS, Canada
- College of Fisheries, Southwest University, Beibei, Chongqing, 400715, China
| | | |
Collapse
|
9
|
Gabriele F, Palerma M, Ippoliti R, Angelucci F, Pitari G, Ardini M. Recent Advances on Affibody- and DARPin-Conjugated Nanomaterials in Cancer Therapy. Int J Mol Sci 2023; 24:ijms24108680. [PMID: 37240041 DOI: 10.3390/ijms24108680] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Affibodies and designed ankyrin repeat proteins (DARPins) are synthetic proteins originally derived from the Staphylococcus aureus virulence factor protein A and the human ankyrin repeat proteins, respectively. The use of these molecules in healthcare has been recently proposed as they are endowed with biochemical and biophysical features heavily demanded to target and fight diseases, as they have a strong binding affinity, solubility, small size, multiple functionalization sites, biocompatibility, and are easy to produce; furthermore, impressive chemical and thermal stability can be achieved. especially when using affibodies. In this sense, several examples reporting on affibodies and DARPins conjugated to nanomaterials have been published, demonstrating their suitability and feasibility in nanomedicine for cancer therapy. This minireview provides a survey of the most recent studies describing affibody- and DARPin-conjugated zero-dimensional nanomaterials, including inorganic, organic, and biological nanoparticles, nanorods, quantum dots, liposomes, and protein- and DNA-based assemblies for targeted cancer therapy in vitro and in vivo.
Collapse
Affiliation(s)
- Federica Gabriele
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Marta Palerma
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Francesco Angelucci
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Giuseppina Pitari
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Matteo Ardini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
10
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [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
|
11
|
Marni R, Malla M, Chakraborty A, Malla R. Proteomic profiling and ROC analysis identify CD151 and ELAVL1 as potential therapy response markers for the antiviral drug in resistant TNBC. Life Sci 2023; 320:121534. [PMID: 36889667 DOI: 10.1016/j.lfs.2023.121534] [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: 12/12/2022] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023]
Abstract
Triple-negative breast cancer is high heterogeneous, aggressive, and metastatic with poor prognosis. Despite of advances in targeted therapies, TNBC has been reported to cause high morbidity and mortality. A rare subpopulation within the tumor microenvironment organized into a hierarchy of cancer stem cells is responsible for therapy resistance and tumor recurrence. Repurposing of antiviral drugs for cancer treatment is gaining momentum due to reduced cost, labour, and research time, but limited due to lack of prognostic, and predictive markers. The present study investigates proteomic profiling and ROC analysis to identify CD151 and ELAVL1 as potential therapy response markers for the antiviral drug 2-thio-6-azauridine (TAU) in resistant TNBC. The stemness of MDA-MB 231 and MDA-MD 468 adherent cells was enriched by culturing them under non-adherent and non-differentiation conditions. Then, CD151+ subpopulation was isolated and characterized for the enrichment of stemness. This study found that CD151 has overexpressed in stemness enriched subpopulations, and also showed CD44 high and CD24 low expression along with stem cell-related transcription factors octamer-binding transcription factor 4 (OCT4) and Sex determining Y-box 2 (SOX2). This study also found that TAU induced significant cytotoxicity and genotoxicity in the CD151+TNBC subpopulation and inhibited their proliferation by inducing DNA damage, cell cycle arrest at the G2M phase, and apoptosis. Further, a proteomic profiling study showed that the expression of CD151 along with ELAVL1, an RNA-binding protein, was significantly reduced with TAU treatment. KM plotter showed correlation of CD151 and ELAVL1 gene expression with a poor prognosis of TNBC. ROC analysis predicted and validated CD151 and ELAVL1 as best therapy response marker for TAU in TNBC. These findings provide new insight into repurposing antiviral drug TAU for treatment of metastatic and drug resistant TNBC.
Collapse
Affiliation(s)
- Rakshmitha Marni
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, A.P., India
| | - Manas Malla
- Department of Computer Science and Engineering, GITAM School of Technology, GITAM (Deemed to be University), Visakhapatnam 530045, A.P., India
| | | | - RamaRao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, A.P., India.
| |
Collapse
|
12
|
Fàbrega C, Clua A, Eritja R, Aviñó A. Oligonucleotides Carrying Nucleoside Antimetabolites as Potential Prodrugs. Curr Med Chem 2023; 30:1304-1319. [PMID: 34844535 PMCID: PMC11497139 DOI: 10.2174/0929867328666211129124039] [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: 07/12/2021] [Revised: 09/07/2021] [Accepted: 09/27/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Nucleoside and nucleobase antimetabolites are an important class of chemotherapeutic agents for the treatment of cancer as well as other diseases. INTRODUCTION In order to avoid undesirable side effects, several prodrug strategies have been developed. In the present review, we describe a relatively unknown strategy that consists of using oligonucleotides modified with nucleoside antimetabolites as prodrugs. METHODS The active nucleotides are generated by enzymatic degradation once incorporated into cells. This strategy has attracted large interest and is widely utilized at present due to the continuous developments made in therapeutic oligonucleotides and the recent advances in nanomaterials and nanomedicine. RESULTS A large research effort was made mainly in the improvement of the antiproliferative properties of nucleoside homopolymers, but recently, chemically modified aptamers, antisense oligonucleotides and/or siRNA carrying antiproliferative nucleotides have demonstrated a great potential due to the synergetic effect of both therapeutic entities. In addition, DNA nanostructures with interesting properties have been built to combine antimetabolites and enhancers of cellular uptake in the same scaffold. Finally, protein nanoparticles functionalized with receptor-binders and antiproliferative oligomers represent a new avenue for a more effective treatment in cancer therapy. CONCLUSION It is expected that oligonucleotides carrying nucleoside antimetabolites will be considered as potential drugs in the near future for biomedical applications.
Collapse
Affiliation(s)
- Carme Fàbrega
- Institute for Advanced Chemistry of Catalonia (IQAC), Spanish National Research Council (CSIC), Barcelona, Spain
- Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-08034 Barcelona, Spain
| | - Anna Clua
- Institute for Advanced Chemistry of Catalonia (IQAC), Spanish National Research Council (CSIC), Barcelona, Spain
- Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-08034 Barcelona, Spain
| | - Ramon Eritja
- Institute for Advanced Chemistry of Catalonia (IQAC), Spanish National Research Council (CSIC), Barcelona, Spain
- Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-08034 Barcelona, Spain
| | - Anna Aviñó
- Institute for Advanced Chemistry of Catalonia (IQAC), Spanish National Research Council (CSIC), Barcelona, Spain
- Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), E-08034 Barcelona, Spain
| |
Collapse
|
13
|
Lin Y, Li Q, Wang L, Guo Q, Liu S, Zhu S, Sun Y, Fan Y, Sun Y, Li H, Tian X, Luo D, Shi S. Advances in regenerative medicine applications of tetrahedral framework nucleic acid-based nanomaterials: an expert consensus recommendation. Int J Oral Sci 2022; 14:51. [PMID: 36316311 PMCID: PMC9622686 DOI: 10.1038/s41368-022-00199-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/09/2022] [Accepted: 08/19/2022] [Indexed: 01/18/2023] Open
Abstract
With the emergence of DNA nanotechnology in the 1980s, self-assembled DNA nanostructures have attracted considerable attention worldwide due to their inherent biocompatibility, unsurpassed programmability, and versatile functions. Especially promising nanostructures are tetrahedral framework nucleic acids (tFNAs), first proposed by Turberfield with the use of a one-step annealing approach. Benefiting from their various merits, such as simple synthesis, high reproducibility, structural stability, cellular internalization, tissue permeability, and editable functionality, tFNAs have been widely applied in the biomedical field as three-dimensional DNA nanomaterials. Surprisingly, tFNAs exhibit positive effects on cellular biological behaviors and tissue regeneration, which may be used to treat inflammatory and degenerative diseases. According to their intended application and carrying capacity, tFNAs could carry functional nucleic acids or therapeutic molecules through extended sequences, sticky-end hybridization, intercalation, and encapsulation based on the Watson and Crick principle. Additionally, dynamic tFNAs also have potential applications in controlled and targeted therapies. This review summarized the latest progress in pure/modified/dynamic tFNAs and demonstrated their regenerative medicine applications. These applications include promoting the regeneration of the bone, cartilage, nerve, skin, vasculature, or muscle and treating diseases such as bone defects, neurological disorders, joint-related inflammatory diseases, periodontitis, and immune diseases.
Collapse
Affiliation(s)
- Yunfeng Lin
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qian Li
- grid.16821.3c0000 0004 0368 8293School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lihua Wang
- grid.458506.a0000 0004 0497 0637The Interdisciplinary Research Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Zhangjiang Laboratory, Shanghai, China
| | - Quanyi Guo
- grid.488137.10000 0001 2267 2324Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, China
| | - Shuyun Liu
- grid.488137.10000 0001 2267 2324Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, China
| | - Shihui Zhu
- grid.73113.370000 0004 0369 1660Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yu Sun
- grid.73113.370000 0004 0369 1660Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yujiang Fan
- grid.13291.380000 0001 0807 1581National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Yong Sun
- grid.13291.380000 0001 0807 1581College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Haihang Li
- Jiangsu Trautec Medical Technology Company Limited, Changzhou, China
| | - Xudong Tian
- Jiangsu Trautec Medical Technology Company Limited, Changzhou, China
| | - Delun Luo
- Chengdu Jingrunze Gene Technology Company Limited, Chengdu, China
| | - Sirong Shi
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Zhang F, Zhang C, Fu S, Liu H, Han M, Fan X, Zhang H, Li W. Amphiphilic Cationic Peptide-Coated PHA Nanosphere as an Efficient Vector for Multiple-Drug Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3024. [PMID: 36080060 PMCID: PMC9457696 DOI: 10.3390/nano12173024] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/08/2022] [Accepted: 08/26/2022] [Indexed: 06/15/2023]
Abstract
Amphiphilic core-shell (ACS) nanoparticles are gaining increasing research interest for multi-drug delivery in cancer therapy. In this work, a new cationic peptide-coated PHA nanosphere was prepared by self-assembly of a hydrophobic core of biodegradable poly (3-hydroxybutyrate-co-3-hydroxyhexanoate) (PHBHHx) and a hydrophilic shell of fusion proteins of PHA granule-associated protein (PhaP) and cationic peptide RALA through a strong hydrophobic effect. The hydrophobic drug curcumin (Cur) was encapsulated in PHBHHx nanoparticles. The chemotherapy drug 5-fluorouracil (5-FU) was administered in the form of its metabolite oligomeric 5-fluorodeoxyuridine (FUdR). Fifteen consecutive FUdR (FUdR15S) were adsorbed on the surface of PHBHHx nanoparticles by electrostatic interaction with RALA to form Cur@PHBX-PR/FUdR15S. Such amphiphilic cationic nanospheres had 88.3% EE of Cur and the drug loading of Cur and FUdR were 7.8% and 12.1%. The dual-drug-loaded nanospheres showed a time-differential release of Cur and FUdR. In addition, Cur@PHBX-PR/FUdR15S exhibited excellent anticancer activity and played a vital role in promoting the synergistic effect of FUdR and Cur in gastric cancer cells. The exploration of antitumor mechanisms demonstrated that Cur improved the activity of apoptosis-related proteins and cancer cells sensitized to FUdR. This amphiphilic core-shell system can serve as a general platform for sequential delivery of multiple drugs to treat several cancer cells.
Collapse
Affiliation(s)
- Fanghua Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
- Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, School of Science, Tianjin University, Tianjin 300072, China
| | - Chao Zhang
- Department of Life Science, Hengshui University, Hengshui 053000, China
| | - Shuangqing Fu
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Huandi Liu
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Mengnan Han
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Xueyu Fan
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Honglei Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Wei Li
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| |
Collapse
|
15
|
Ping L, Ruxian J, Mengping Z, Pei J, Zhuoya L, Guosheng L, Zhenyu W, Hailei W. Whole-cell biosynthesis of cytarabine by an unnecessary protein-reduced Escherichia coli that coexpresses purine and uracil phosphorylase. Biotechnol Bioeng 2022; 119:1768-1780. [PMID: 35383880 DOI: 10.1002/bit.28098] [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: 02/12/2022] [Revised: 03/18/2022] [Accepted: 03/25/2022] [Indexed: 11/10/2022]
Abstract
Currently, whole-cell catalysts face challenges due to the complexity of reaction systems, although they have a cost advantage over pure enzymes. In this work, cytarabine was synthesized by purified purine phosphorylase 1 (PNP1) and uracil phosphorylase (UP), and the conversion of cytarabine from adenine arabinoside reached 72.3±4.3%. However, the synthesis was unsuccessful by whole-cell catalysis due to interference from unnecessary proteins (UNPs) in cells. Thus, we carried out a large-scale gene editing involving 377 genes in the genome of Escherichia coli to reduce the negative effect of UNPs on substrate conversion and cytarabine production. Finally, the PNP1 and UP activities of the obtained mutant were increased significantly compared with the parental strain, and more importantly, the conversion rate of cytarabine by whole-cell catalysis reached 67.4±2.5%. The lack of 148 proteins and down-regulation of 783 proteins caused by gene editing were equivalent to partial purification of the enzymes within cells, and thus, we provided inspiration to solve the problem caused by UNP interference, which is ubiquitous in the field of whole-cell catalysis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Li Ping
- Henan Engineering Laboratory for Bioconversion Technology of Functional Microbes,College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Jing Ruxian
- Henan Engineering Laboratory for Bioconversion Technology of Functional Microbes,College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Zhou Mengping
- Henan Engineering Laboratory for Bioconversion Technology of Functional Microbes,College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Jia Pei
- Henan Engineering Laboratory for Bioconversion Technology of Functional Microbes,College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Li Zhuoya
- Henan Engineering Laboratory for Bioconversion Technology of Functional Microbes,College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Liu Guosheng
- Henan Engineering Laboratory for Bioconversion Technology of Functional Microbes,College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Wang Zhenyu
- Henan Engineering Laboratory for Bioconversion Technology of Functional Microbes,College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Wang Hailei
- Henan Engineering Laboratory for Bioconversion Technology of Functional Microbes,College of Life Science, Henan Normal University, Xinxiang, 453007, China.,Advanced Environmental Biotechnology Center, Nanyang Technological University, Singapore, 637141, Singapore
| |
Collapse
|
16
|
Zhang C, Fu S, Zhang F, Han M, Wang X, Du J, Zhang H, Li W. Affibody Modified G-quadruplex DNA Micelles Incorporating Polymeric 5-Fluorodeoxyuridine for Targeted Delivery of Curcumin to Enhance Synergetic Therapy of HER2 Positive Gastric Cancer. NANOMATERIALS 2022; 12:nano12040696. [PMID: 35215023 PMCID: PMC8879187 DOI: 10.3390/nano12040696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/12/2022]
Abstract
Combination chemotherapy is emerging as an important strategy for cancer treatment with decreased side effects. However, chemotherapeutic drugs with different solubility are not easy to realize co-delivery in traditional nanocarriers. Herein, an affibody modified G-quadruplex DNA micellar prodrug (affi-F/GQs) of hydrophilic 5-fluorodeoxyuridine (FUdR) by integrating polymeric FUdRs into DNA strands is developed for the first time. To achieve synergistic efficacy with hydrophobic drugs, curcumin (Cur) is co-loaded into affi-F/GQs micelles to prepare the dual drug-loaded DNA micelles (Cur@affi-F/GQs), in which affibody is employed as a targeting moiety to facilitate HER2 receptor-mediated uptake. Cur@affi-F/GQs have a small size of approximately 130 nm and exhibit excellent stability. The system co-delivers FUdR and Cur in a ratiometric manner, and the drug loading rates are 21.1% and 5.6%, respectively. Compared with the physical combination of FUdR and Cur, Cur@affi-F/GQs show higher cytotoxicity and greater synergistic effect on HER2 positive gastric cancer N87 cells. Surprisingly, Cur@affi-F/GQs significantly enhance the expression and activity of apoptosis-associated proteins in Bcl-2/Bax-caspase 8, 9-caspase 3 apoptotic pathway, which is the main factor in the death of tumor cells induced by FUdR. Overall, this nanoencapsulation is a promising candidate for the targeted co-delivery of drugs with significant differences in solubility.
Collapse
Affiliation(s)
- Chao Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province-Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China; (C.Z.); (S.F.); (F.Z.); (M.H.); (X.W.); (J.D.)
- Department of Life Science, Hengshui University, Hengshui 053000, China
| | - Shuangqing Fu
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province-Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China; (C.Z.); (S.F.); (F.Z.); (M.H.); (X.W.); (J.D.)
| | - Fanghua Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province-Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China; (C.Z.); (S.F.); (F.Z.); (M.H.); (X.W.); (J.D.)
| | - Mengnan Han
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province-Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China; (C.Z.); (S.F.); (F.Z.); (M.H.); (X.W.); (J.D.)
| | - Xuming Wang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province-Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China; (C.Z.); (S.F.); (F.Z.); (M.H.); (X.W.); (J.D.)
| | - Jie Du
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province-Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China; (C.Z.); (S.F.); (F.Z.); (M.H.); (X.W.); (J.D.)
| | - Honglei Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province-Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China; (C.Z.); (S.F.); (F.Z.); (M.H.); (X.W.); (J.D.)
- Correspondence: (H.Z.); (W.L.)
| | - Wei Li
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province-Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China; (C.Z.); (S.F.); (F.Z.); (M.H.); (X.W.); (J.D.)
- Correspondence: (H.Z.); (W.L.)
| |
Collapse
|
17
|
Shipunova VO, Deyev SM. Artificial Scaffold Polypeptides As an Efficient Tool for the Targeted Delivery of Nanostructures In Vitro and In Vivo. Acta Naturae 2022; 14:54-72. [PMID: 35441046 PMCID: PMC9013437 DOI: 10.32607/actanaturae.11545] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/20/2021] [Indexed: 12/22/2022] Open
Abstract
The use of traditional tools for the targeted delivery of nanostructures, such as antibodies, transferrin, lectins, or aptamers, often leads to an entire range of undesirable effects. The large size of antibodies often does not allow one to reach the required number of molecules on the surface of nanostructures during modification, and the constant domains of heavy chains, due to their effector functions, can induce phagocytosis. In the recent two decades, targeted polypeptide scaffold molecules of a non-immunoglobulin nature, antibody mimetics, have emerged as much more effective targeting tools. They are small in size (3-20 kDa), possess high affinity (from subnano- to femtomolar binding constants), low immunogenicity, and exceptional thermodynamic stability. These molecules can be effectively produced in bacterial cells, and, using genetic engineering manipulations, it is possible to create multispecific fusion proteins for the targeting of nanoparticles to cells with a given molecular portrait, which makes scaffold polypeptides an optimal tool for theranostics.
Collapse
Affiliation(s)
- V. O. Shipunova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| | - S. M. Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997 Russia
| |
Collapse
|
18
|
Wang Y, Wang Z, Wu X, Liu S, Liu F, Jiang Q, Ding B. DNA Nanodevices: from Mechanical Motions to Biomedical Applications. Curr Top Med Chem 2021; 22:640-651. [PMID: 34749612 DOI: 10.2174/1568026621666211105100240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/11/2021] [Accepted: 10/11/2021] [Indexed: 11/22/2022]
Abstract
Inspired by molecular machines in nature, artificial nanodevices have been designed to realize various biomedical functions. Self-assembled deoxyribonucleic acid (DNA) nanostructures that feature designed geometries, excellent spatial accuracy, nanoscale addressability and marked biocompatibility provide an attractive candidate for constructing dynamic nanodevices with biomarker-targeting and stimuli-responsiveness for biomedical applications. Here, a summary of typical construction strategies of DNA nanodevices and their operating mechanisms are presented. We also introduced recent advances in employing DNA nanodevices as platforms for biosensing and intelligent drug delivery. Finally, the broad prospects and main challenges of the DNA nanodevices in biomedical applications are discussed.
Collapse
Affiliation(s)
- Yiming Wang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing. China
| | - Zhaoran Wang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing. China
| | - Xiaohui Wu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing. China
| | - Shaoli Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing. China
| | - Fengsong Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing. China
| | - Qiao Jiang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing. China
| | - Baoquan Ding
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for NanoScience and Technology, Beijing. China
| |
Collapse
|
19
|
Seitz I, Shaukat A, Nurmi K, Ijäs H, Hirvonen J, Santos HA, Kostiainen MA, Linko V. Prospective Cancer Therapies Using Stimuli-Responsive DNA Nanostructures. Macromol Biosci 2021; 21:e2100272. [PMID: 34614301 DOI: 10.1002/mabi.202100272] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/28/2021] [Indexed: 11/08/2022]
Abstract
Nanostructures based on DNA self-assembly present an innovative way to address the increasing need for target-specific delivery of therapeutic molecules. Currently, most of the chemotherapeutics being used in clinical practice have undesired and exceedingly high off-target toxicity. This is a challenge in particular for small molecules, and hence, developing robust and effective methods to lower these side effects and enhance the antitumor activity is of paramount importance. Prospectively, these issues could be tackled with the help of DNA nanotechnology, which provides a route for the fabrication of custom, biocompatible, and multimodal structures, which can, to some extent, resist nuclease degradation and survive in the cellular environment. Similar to widely employed liposomal products, the DNA nanostructures (DNs) are loaded with selected drugs, and then by employing a specific stimulus, the payload can be released at its target region. This review explores several strategies and triggers to achieve targeted delivery of DNs. Notably, different modalities are explained through which DNs can interact with their respective targets as well as how structural changes triggered by external stimuli can be used to achieve the display or release of the cargo. Furthermore, the prospects and challenges of this technology are highlighted.
Collapse
Affiliation(s)
- Iris Seitz
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, P.O. Box 16100, Aalto, 00076, Finland
| | - Ahmed Shaukat
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, P.O. Box 16100, Aalto, 00076, Finland
| | - Kurt Nurmi
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| | - Heini Ijäs
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, P.O. Box 16100, Aalto, 00076, Finland.,Nanoscience Center, Department of Biological and Environmental Science, University of Jyväskylä, P.O. Box 35, Jyväskylä, 40014, Finland
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland.,Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, University Medical Center Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Mauri A Kostiainen
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, P.O. Box 16100, Aalto, 00076, Finland.,HYBER Centre, Department of Applied Physics, Aalto University, P.O. Box 15100, Aalto, 00076, Finland
| | - Veikko Linko
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, P.O. Box 16100, Aalto, 00076, Finland.,HYBER Centre, Department of Applied Physics, Aalto University, P.O. Box 15100, Aalto, 00076, Finland
| |
Collapse
|
20
|
Rai A, Noor S, Ahmad SI, Alajmi MF, Hussain A, Abbas H, Hasan GM. Recent Advances and Implication of Bioengineered Nanomaterials in Cancer Theranostics. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:91. [PMID: 33494239 PMCID: PMC7909769 DOI: 10.3390/medicina57020091] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/28/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023]
Abstract
Cancer is one of the most common causes of death and affects millions of lives every year. In addition to non-infectious carcinogens, infectious agents contribute significantly to increased incidence of several cancers. Several therapeutic techniques have been used for the treatment of such cancers. Recently, nanotechnology has emerged to advance the diagnosis, imaging, and therapeutics of various cancer types. Nanomaterials have multiple advantages over other materials due to their small size and high surface area, which allow retention and controlled drug release to improve the anti-cancer property. Most cancer therapies have been known to damage healthy cells due to poor specificity, which can be avoided by using nanosized particles. Nanomaterials can be combined with various types of biomaterials to make it less toxic and improve its biocompatibility. Based on these properties, several nanomaterials have been developed which possess excellent anti-cancer efficacy potential and improved diagnosis. This review presents the latest update on novel nanomaterials used to improve the diagnostic and therapeutic of pathogen-associated and non-pathogenic cancers. We further highlighted mechanistic insights into their mode of action, improved features, and limitations.
Collapse
Affiliation(s)
- Ayushi Rai
- Department of Nanoscience, Central University of Gujarat, Sector 29, Gandhinagar 382030, India;
| | - Saba Noor
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India;
| | - Syed Ishraque Ahmad
- Department of Chemistry, Zakir Husain Delhi College, University of Delhi, New Delhi 110002, India;
| | - Mohamed F. Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.F.A.); (A.H.)
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.F.A.); (A.H.)
| | - Hashim Abbas
- Department of Medicine, Nottingham University Hospitals, NHS Trust, Nottingham NG7 2UH, UK;
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
21
|
Zhang C, Zhang F, Han M, Wang X, Du J, Zhang H, Li W. Co-delivery of 5-fluorodeoxyuridine and doxorubicin via gold nanoparticle equipped with affibody-DNA hybrid strands for targeted synergistic chemotherapy of HER2 overexpressing breast cancer. Sci Rep 2020; 10:22015. [PMID: 33328545 PMCID: PMC7745031 DOI: 10.1038/s41598-020-79125-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 11/23/2020] [Indexed: 01/12/2023] Open
Abstract
Combination chemotherapy is still of great importance as part of the standard clinical care for patients with HER2 positive breast cancer. As an attractive component, gold nanoparticles (AuNPs) have been extensively studied as biosafety nanomaterials, but they are rarely explored as drug nanocarriers for targeted co-delivery of multiple chemotherapeutics. Herein, a novel affibody-DNA hybrid strands modified AuNPs were fabricated for co-loading nucleoside analogue (5-fluorodeoxyuridine, FUdR) and anthracycline (doxorubicin, Dox). FUdRs were integrated into DNA hybrid strands decorated on AuNPs by DNA solid phase synthesis, and Dox molecules were intercalated into their duplex regions. Affibody molecules coupled to the DNA hybrid strands were distributed the surface of AuNPs, giving them targeting for HER2. The new dual-drug-containing affibody-DNA-AuNPs (Dox@affi-F/AuNPs) owned compact and stable spherical nanostructures, and precise drug loading. Cytotoxicity tests demonstrated that these nanoparticles caused a higher inhibition in HER2 overexpressing breast cancer cells, and showed better synergistic antitumor activity than simple mixture of the two drugs. The related mechanistic studies proved that Dox@affi-F/AuNPs achieved a remarkable combined antitumor activity of Dox and FUdR by promoting more cells to enter apoptosis pathway. Our work provided a nanomedicine platform for targeted co-delivery of nucleoside analog therapeutics and anthracycline anticancer drugs to achieve synergistic treatment of HER2+ cancer.
Collapse
Affiliation(s)
- Chao Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Fanghua Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Mengnan Han
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Xuming Wang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Jie Du
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Honglei Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China.
| | - Wei Li
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China.
| |
Collapse
|
22
|
Yu L, Wang F, Tai M, Li J, Gong S, Zhou Z, Yin X, Gu X, Li C. 6H2L, a novel synthetic derivative of bifendate, induces apoptosis in hepatoma cells via mitochondrial and MAPK pathway. Eur J Pharmacol 2020; 882:173299. [PMID: 32589884 DOI: 10.1016/j.ejphar.2020.173299] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related death worldwide. Our previous study indicated that 6H2L, a novel synthetic bifendate derivative, shows multidrug resistance reversal activity, while its antitumor effect has not been revealed. Here, the potent antitumor effects of 6H2L on hepatoma cells both in vitro and in vivo were investigated. 6H2L inhibited cell viability of HepG2 and SMMC-7721 cells with less sensitivity to normal human liver L-02 cells. 6H2L induced apoptosis in hepatoma cells. It upregulated Bax expression, while simultaneously decreasing Bcl-2 expression. Further elucidation of the mechanism revealed that 6H2L induced mitochondrial dysfunction, with transmitochondrial membrane potential collapse and cytochrome c release, which activated caspase-9 and caspase-3 and subsequently cleaved PARP, suggesting that 6H2L induced apoptosis via triggering mitochondrial pathway. Moreover, 6H2L decreased the phosphorylation of ERK1/2, whereas it increased the expression of p-JNK and p-p38. Then, specific inhibitors of the mitogen-activated protein kinase (MAPK) pathway were employed to confirm the roles of the MAPK pathway in the apoptosis-inducing effects of 6H2L. Additionally, 6H2L obviously inhibited the tumor growth in H22-bearing ICR mice. Meanwhile, 6H2L remarkably up-regulated Bax while suppressing Bcl-2 in tumors. Importantly, neither significant weight loss, white blood cell (WBC) count, nor histopathological abnormalities of major organs were observed in the mice receiving 6H2L treatment, indicating that 6H2L exerted strong anticancer activities with low toxicity in vivo. In contrast, fluorouracil inhibited tumor growth with significant decreased body weight and WBC count. Taken together, these results suggested 6H2L is a potential therapeutic candidate for HCC.
Collapse
Affiliation(s)
- Lirong Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Fan Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Mengying Tai
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Juan Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Shuyuan Gong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Zhengwei Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Xiaoke Gu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, People's Republic of China.
| |
Collapse
|
23
|
Zhang F, Yin J, Zhang C, Han M, Wang X, Fu S, Du J, Zhang H, Li W. Affibody-Conjugated RALA Polymers Delivering Oligomeric 5-Fluorodeoxyuridine for Targeted Therapy of HER2 Overexpressing Gastric Cancer. Macromol Biosci 2020; 20:e2000083. [PMID: 32558229 DOI: 10.1002/mabi.202000083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/30/2020] [Indexed: 11/10/2022]
Abstract
Affibody-conjugated RALA (affi-RA) are designed for delivering oligomeric 5-fluorodeoxyuridine (FUdR, metabolite of 5-FU) strand to raise the selectivity of 5-fluorouracil (5-FU), decrease its toxicity and improve its suboptimal therapeutic efficacy. The nanodrugs, FUdR@affi-RA, are spontaneously assembled by electrostatic interaction between positively charged affi-RA and negatively charged FUdR15 -strands (15 consecutive FUdR). FUdR@affi-RA exhibits excellent stability under simulated physiological conditions. Compared with free FUdR, FUdR@affi-RA shows excellent targeting and higher cytotoxicity in human epidermal growth factor receptor 2 (HER2) overexpressing gastric cancer N87 cells. Moreover, the anticancer mechanism studies reveal that FUdR@affi-RA enhances the expression and activity of apoptosis-associated proteins in the Bcl-2/Bax-caspase 8,9-caspase 3 apoptotic pathway induced by FUdR. This study indicates that the fusion vector, affi-RA, presents a promising delivery system platform for nucleoside analogue drugs and provides a new strategy for the development of therapeutics of cancer treatment.
Collapse
Affiliation(s)
- Fanghua Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Jiwei Yin
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Chao Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Mengnan Han
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Xuming Wang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Shuangqing Fu
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Jie Du
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Honglei Zhang
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| | - Wei Li
- College of Chemistry and Environmental Science, Key Laboratory of Chemical Biology of Hebei Province, Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, China
| |
Collapse
|